EP4257139A2 - Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds - Google Patents
Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds Download PDFInfo
- Publication number
- EP4257139A2 EP4257139A2 EP23166953.2A EP23166953A EP4257139A2 EP 4257139 A2 EP4257139 A2 EP 4257139A2 EP 23166953 A EP23166953 A EP 23166953A EP 4257139 A2 EP4257139 A2 EP 4257139A2
- Authority
- EP
- European Patent Office
- Prior art keywords
- muscle
- present disclosure
- composition
- group
- chemical formula
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 150000001875 compounds Chemical class 0.000 title claims abstract description 69
- 239000000203 mixture Substances 0.000 title claims abstract description 65
- 208000021642 Muscular disease Diseases 0.000 title claims abstract description 46
- 229940124530 sulfonamide Drugs 0.000 title claims abstract description 23
- 150000003456 sulfonamides Chemical class 0.000 title claims abstract description 18
- 150000003839 salts Chemical class 0.000 claims abstract description 33
- 235000013305 food Nutrition 0.000 claims abstract description 29
- 239000000126 substance Substances 0.000 claims description 66
- 208000001076 sarcopenia Diseases 0.000 claims description 33
- 239000008194 pharmaceutical composition Substances 0.000 claims description 29
- 125000001424 substituent group Chemical group 0.000 claims description 22
- 238000011282 treatment Methods 0.000 claims description 22
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 20
- 206010028289 Muscle atrophy Diseases 0.000 claims description 19
- 125000001072 heteroaryl group Chemical group 0.000 claims description 18
- 125000003118 aryl group Chemical group 0.000 claims description 17
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 claims description 14
- 125000006274 (C1-C3)alkoxy group Chemical group 0.000 claims description 14
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 14
- 201000000585 muscular atrophy Diseases 0.000 claims description 13
- 230000002265 prevention Effects 0.000 claims description 10
- WTKZEGDFNFYCGP-UHFFFAOYSA-N Pyrazole Chemical compound C=1C=NNC=1 WTKZEGDFNFYCGP-UHFFFAOYSA-N 0.000 claims description 9
- 230000006872 improvement Effects 0.000 claims description 9
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 claims description 7
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 claims description 7
- VLLMWSRANPNYQX-UHFFFAOYSA-N thiadiazole Chemical compound C1=CSN=N1.C1=CSN=N1 VLLMWSRANPNYQX-UHFFFAOYSA-N 0.000 claims description 7
- 125000000218 acetic acid group Chemical group C(C)(=O)* 0.000 claims description 6
- 239000001257 hydrogen Substances 0.000 claims description 6
- 229910052739 hydrogen Inorganic materials 0.000 claims description 6
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 6
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical compound C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 claims description 5
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 5
- 206010006895 Cachexia Diseases 0.000 claims description 4
- 206010028372 Muscular weakness Diseases 0.000 claims description 4
- 201000006938 muscular dystrophy Diseases 0.000 claims description 4
- 230000036473 myasthenia Effects 0.000 claims description 4
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims 3
- 210000003205 muscle Anatomy 0.000 abstract description 66
- 238000011084 recovery Methods 0.000 abstract description 34
- 230000004069 differentiation Effects 0.000 abstract description 27
- 230000001965 increasing effect Effects 0.000 abstract description 23
- 230000014509 gene expression Effects 0.000 abstract description 19
- 102100036878 PHD finger protein 20 Human genes 0.000 abstract description 18
- 101001071230 Homo sapiens PHD finger protein 20 Proteins 0.000 abstract description 17
- 210000003098 myoblast Anatomy 0.000 abstract description 13
- 239000003814 drug Substances 0.000 abstract description 12
- 230000005764 inhibitory process Effects 0.000 abstract description 9
- 230000004220 muscle function Effects 0.000 abstract description 9
- 230000009756 muscle regeneration Effects 0.000 abstract description 7
- 230000001105 regulatory effect Effects 0.000 abstract description 4
- 230000006870 function Effects 0.000 abstract description 3
- 229940124597 therapeutic agent Drugs 0.000 abstract description 3
- 208000029578 Muscle disease Diseases 0.000 abstract description 2
- 210000001087 myotubule Anatomy 0.000 abstract 1
- 241000699670 Mus sp. Species 0.000 description 87
- 229960002135 sulfadimidine Drugs 0.000 description 74
- ASWVTGNCAZCNNR-UHFFFAOYSA-N sulfamethazine Chemical compound CC1=CC(C)=NC(NS(=O)(=O)C=2C=CC(N)=CC=2)=N1 ASWVTGNCAZCNNR-UHFFFAOYSA-N 0.000 description 71
- 230000001988 toxicity Effects 0.000 description 38
- 231100000419 toxicity Toxicity 0.000 description 38
- NCEXYHBECQHGNR-QZQOTICOSA-N sulfasalazine Chemical compound C1=C(O)C(C(=O)O)=CC(\N=N\C=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-QZQOTICOSA-N 0.000 description 37
- UETNIIAIRMUTSM-UHFFFAOYSA-N Jacareubin Natural products CC1(C)OC2=CC3Oc4c(O)c(O)ccc4C(=O)C3C(=C2C=C1)O UETNIIAIRMUTSM-UHFFFAOYSA-N 0.000 description 36
- 229960001940 sulfasalazine Drugs 0.000 description 36
- NCEXYHBECQHGNR-UHFFFAOYSA-N sulfasalazine Natural products C1=C(O)C(C(=O)O)=CC(N=NC=2C=CC(=CC=2)S(=O)(=O)NC=2N=CC=CC=2)=C1 NCEXYHBECQHGNR-UHFFFAOYSA-N 0.000 description 36
- 210000004027 cell Anatomy 0.000 description 35
- 235000002639 sodium chloride Nutrition 0.000 description 35
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 33
- 230000000694 effects Effects 0.000 description 32
- 239000002953 phosphate buffered saline Substances 0.000 description 32
- 238000010172 mouse model Methods 0.000 description 30
- 230000037396 body weight Effects 0.000 description 29
- 241000699666 Mus <mouse, genus> Species 0.000 description 28
- 238000012790 confirmation Methods 0.000 description 19
- 238000002347 injection Methods 0.000 description 15
- 239000007924 injection Substances 0.000 description 15
- 208000027418 Wounds and injury Diseases 0.000 description 14
- 230000007423 decrease Effects 0.000 description 14
- 238000013537 high throughput screening Methods 0.000 description 14
- 230000032683 aging Effects 0.000 description 13
- 238000002474 experimental method Methods 0.000 description 13
- 210000002414 leg Anatomy 0.000 description 13
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 12
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 11
- 230000006378 damage Effects 0.000 description 11
- 229960003722 doxycycline Drugs 0.000 description 11
- XQTWDDCIUJNLTR-CVHRZJFOSA-N doxycycline monohydrate Chemical compound O.O=C1C2=C(O)C=CC=C2[C@H](C)[C@@H]2C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H](N(C)C)[C@@H]1[C@H]2O XQTWDDCIUJNLTR-CVHRZJFOSA-N 0.000 description 11
- 208000014674 injury Diseases 0.000 description 11
- 238000005259 measurement Methods 0.000 description 11
- 238000000034 method Methods 0.000 description 11
- 238000002360 preparation method Methods 0.000 description 11
- 238000010586 diagram Methods 0.000 description 10
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- -1 methoxy, ethoxy Chemical group 0.000 description 10
- 210000000663 muscle cell Anatomy 0.000 description 10
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 9
- 235000005911 diet Nutrition 0.000 description 9
- 230000037213 diet Effects 0.000 description 9
- 229940079593 drug Drugs 0.000 description 9
- 230000001976 improved effect Effects 0.000 description 9
- 238000004020 luminiscence type Methods 0.000 description 9
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 9
- 102100031142 Transcriptional repressor protein YY1 Human genes 0.000 description 8
- 239000004480 active ingredient Substances 0.000 description 8
- 230000008859 change Effects 0.000 description 8
- 230000007850 degeneration Effects 0.000 description 8
- 230000036541 health Effects 0.000 description 8
- 235000009200 high fat diet Nutrition 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 7
- 229920002472 Starch Chemical class 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 230000002401 inhibitory effect Effects 0.000 description 7
- 210000004185 liver Anatomy 0.000 description 7
- 239000003550 marker Substances 0.000 description 7
- 210000002027 skeletal muscle Anatomy 0.000 description 7
- 239000011780 sodium chloride Substances 0.000 description 7
- 208000024891 symptom Diseases 0.000 description 7
- 244000025254 Cannabis sativa Species 0.000 description 6
- 208000029549 Muscle injury Diseases 0.000 description 6
- 108060008487 Myosin Proteins 0.000 description 6
- 102000003505 Myosin Human genes 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- PJSFRIWCGOHTNF-UHFFFAOYSA-N Sulphormetoxin Chemical compound COC1=NC=NC(NS(=O)(=O)C=2C=CC(N)=CC=2)=C1OC PJSFRIWCGOHTNF-UHFFFAOYSA-N 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- 235000013376 functional food Nutrition 0.000 description 6
- 125000005842 heteroatom Chemical group 0.000 description 6
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 6
- 239000004615 ingredient Substances 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- 239000003755 preservative agent Substances 0.000 description 6
- 235000019698 starch Nutrition 0.000 description 6
- 230000000638 stimulation Effects 0.000 description 6
- SEEPANYCNGTZFQ-UHFFFAOYSA-N sulfadiazine Chemical compound C1=CC(N)=CC=C1S(=O)(=O)NC1=NC=CC=N1 SEEPANYCNGTZFQ-UHFFFAOYSA-N 0.000 description 6
- 229960004306 sulfadiazine Drugs 0.000 description 6
- 229960000973 sulfadimethoxine Drugs 0.000 description 6
- ZZORFUFYDOWNEF-UHFFFAOYSA-N sulfadimethoxine Chemical compound COC1=NC(OC)=CC(NS(=O)(=O)C=2C=CC(N)=CC=2)=N1 ZZORFUFYDOWNEF-UHFFFAOYSA-N 0.000 description 6
- 229960004673 sulfadoxine Drugs 0.000 description 6
- GPTONYMQFTZPKC-UHFFFAOYSA-N sulfamethoxydiazine Chemical compound N1=CC(OC)=CN=C1NS(=O)(=O)C1=CC=C(N)C=C1 GPTONYMQFTZPKC-UHFFFAOYSA-N 0.000 description 6
- 229960002229 sulfametoxydiazine Drugs 0.000 description 6
- 229960004818 sulfaphenazole Drugs 0.000 description 6
- QWCJHSGMANYXCW-UHFFFAOYSA-N sulfaphenazole Chemical compound C1=CC(N)=CC=C1S(=O)(=O)NC1=CC=NN1C1=CC=CC=C1 QWCJHSGMANYXCW-UHFFFAOYSA-N 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- 238000004804 winding Methods 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 5
- 238000013218 HFD mouse model Methods 0.000 description 5
- 239000002671 adjuvant Substances 0.000 description 5
- 239000003674 animal food additive Substances 0.000 description 5
- 210000004369 blood Anatomy 0.000 description 5
- 239000008280 blood Substances 0.000 description 5
- 231100000259 cardiotoxicity Toxicity 0.000 description 5
- 239000013592 cell lysate Substances 0.000 description 5
- 230000001413 cellular effect Effects 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 230000003203 everyday effect Effects 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 210000002216 heart Anatomy 0.000 description 5
- 230000007678 heart toxicity Effects 0.000 description 5
- 238000007490 hematoxylin and eosin (H&E) staining Methods 0.000 description 5
- 231100000304 hepatotoxicity Toxicity 0.000 description 5
- 238000003125 immunofluorescent labeling Methods 0.000 description 5
- 230000007056 liver toxicity Effects 0.000 description 5
- 238000002156 mixing Methods 0.000 description 5
- 231100000417 nephrotoxicity Toxicity 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 239000008107 starch Chemical class 0.000 description 5
- 229960000551 sulfacetamide sodium Drugs 0.000 description 5
- IHCDKJZZFOUARO-UHFFFAOYSA-M sulfacetamide sodium Chemical compound O.[Na+].CC(=O)[N-]S(=O)(=O)C1=CC=C(N)C=C1 IHCDKJZZFOUARO-UHFFFAOYSA-M 0.000 description 5
- 229960005158 sulfamethizole Drugs 0.000 description 5
- VACCAVUAMIDAGB-UHFFFAOYSA-N sulfamethizole Chemical compound S1C(C)=NN=C1NS(=O)(=O)C1=CC=C(N)C=C1 VACCAVUAMIDAGB-UHFFFAOYSA-N 0.000 description 5
- FDDDEECHVMSUSB-UHFFFAOYSA-N sulfanilamide Chemical compound NC1=CC=C(S(N)(=O)=O)C=C1 FDDDEECHVMSUSB-UHFFFAOYSA-N 0.000 description 5
- 229960002211 sulfapyridine Drugs 0.000 description 5
- GECHUMIMRBOMGK-UHFFFAOYSA-N sulfapyridine Chemical compound C1=CC(N)=CC=C1S(=O)(=O)NC1=CC=CC=N1 GECHUMIMRBOMGK-UHFFFAOYSA-N 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 241000251468 Actinopterygii Species 0.000 description 4
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 4
- YLQBMQCUIZJEEH-UHFFFAOYSA-N Furan Chemical compound C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 4
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 4
- 229920001213 Polysorbate 20 Polymers 0.000 description 4
- KAESVJOAVNADME-UHFFFAOYSA-N Pyrrole Chemical compound C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 description 4
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 235000013361 beverage Nutrition 0.000 description 4
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 4
- 238000010276 construction Methods 0.000 description 4
- 235000015872 dietary supplement Nutrition 0.000 description 4
- JBIWCJUYHHGXTC-AKNGSSGZSA-N doxycycline Chemical compound O=C1C2=C(O)C=CC=C2[C@H](C)[C@@H]2C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H](N(C)C)[C@@H]1[C@H]2O JBIWCJUYHHGXTC-AKNGSSGZSA-N 0.000 description 4
- 235000019688 fish Nutrition 0.000 description 4
- 235000003599 food sweetener Nutrition 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- 229960001031 glucose Drugs 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 229910052500 inorganic mineral Inorganic materials 0.000 description 4
- 210000000936 intestine Anatomy 0.000 description 4
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 4
- 210000003734 kidney Anatomy 0.000 description 4
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 4
- 210000004072 lung Anatomy 0.000 description 4
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 4
- 235000010755 mineral Nutrition 0.000 description 4
- 239000011707 mineral Substances 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 235000019198 oils Nutrition 0.000 description 4
- 210000000496 pancreas Anatomy 0.000 description 4
- 238000007911 parenteral administration Methods 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 4
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 4
- 102000004169 proteins and genes Human genes 0.000 description 4
- 108090000623 proteins and genes Proteins 0.000 description 4
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 4
- 230000008929 regeneration Effects 0.000 description 4
- 238000011069 regeneration method Methods 0.000 description 4
- 238000012216 screening Methods 0.000 description 4
- 235000020183 skimmed milk Nutrition 0.000 description 4
- 239000003381 stabilizer Substances 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 239000011550 stock solution Substances 0.000 description 4
- BRBKOPJOKNSWSG-UHFFFAOYSA-N sulfaguanidine Chemical compound NC(=N)NS(=O)(=O)C1=CC=C(N)C=C1 BRBKOPJOKNSWSG-UHFFFAOYSA-N 0.000 description 4
- 229960004257 sulfaguanidine Drugs 0.000 description 4
- 229960001544 sulfathiazole Drugs 0.000 description 4
- JNMRHUJNCSQMMB-UHFFFAOYSA-N sulfathiazole Chemical compound C1=CC(N)=CC=C1S(=O)(=O)NC1=NC=CS1 JNMRHUJNCSQMMB-UHFFFAOYSA-N 0.000 description 4
- 239000003765 sweetening agent Substances 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 239000000080 wetting agent Substances 0.000 description 4
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical class O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 3
- 241000894006 Bacteria Species 0.000 description 3
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 3
- 102000016798 Myosin Type I Human genes 0.000 description 3
- 108010028277 Myosin Type I Proteins 0.000 description 3
- 241000283984 Rodentia Species 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 239000000443 aerosol Substances 0.000 description 3
- 210000003423 ankle Anatomy 0.000 description 3
- 239000003963 antioxidant agent Substances 0.000 description 3
- 235000006708 antioxidants Nutrition 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 229940077731 carbohydrate nutrients Drugs 0.000 description 3
- 150000001720 carbohydrates Chemical class 0.000 description 3
- 235000014633 carbohydrates Nutrition 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 235000013339 cereals Nutrition 0.000 description 3
- 230000000052 comparative effect Effects 0.000 description 3
- 239000012141 concentrate Substances 0.000 description 3
- 235000008504 concentrate Nutrition 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 231100000673 dose–response relationship Toxicity 0.000 description 3
- 238000001647 drug administration Methods 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 239000003995 emulsifying agent Substances 0.000 description 3
- 235000015203 fruit juice Nutrition 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 239000008101 lactose Substances 0.000 description 3
- 229960001375 lactose Drugs 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 210000004165 myocardium Anatomy 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 230000002335 preservative effect Effects 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 235000018102 proteins Nutrition 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 159000000000 sodium salts Chemical class 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 210000002784 stomach Anatomy 0.000 description 3
- 229940101581 sulfasalazine 500 mg Drugs 0.000 description 3
- 239000003826 tablet Substances 0.000 description 3
- 210000001550 testis Anatomy 0.000 description 3
- 235000013343 vitamin Nutrition 0.000 description 3
- 239000011782 vitamin Substances 0.000 description 3
- 229940088594 vitamin Drugs 0.000 description 3
- 229930003231 vitamin Natural products 0.000 description 3
- JYEUMXHLPRZUAT-UHFFFAOYSA-N 1,2,3-triazine Chemical compound C1=CN=NN=C1 JYEUMXHLPRZUAT-UHFFFAOYSA-N 0.000 description 2
- SXAMGRAIZSSWIH-UHFFFAOYSA-N 2-[3-[2-(2,3-dihydro-1H-inden-2-ylamino)pyrimidin-5-yl]-1,2,4-oxadiazol-5-yl]-1-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)ethanone Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C1=NOC(=N1)CC(=O)N1CC2=C(CC1)NN=N2 SXAMGRAIZSSWIH-UHFFFAOYSA-N 0.000 description 2
- BCHZICNRHXRCHY-UHFFFAOYSA-N 2h-oxazine Chemical compound N1OC=CC=C1 BCHZICNRHXRCHY-UHFFFAOYSA-N 0.000 description 2
- AGIJRRREJXSQJR-UHFFFAOYSA-N 2h-thiazine Chemical compound N1SC=CC=C1 AGIJRRREJXSQJR-UHFFFAOYSA-N 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 240000002791 Brassica napus Species 0.000 description 2
- 235000011293 Brassica napus Nutrition 0.000 description 2
- 235000000540 Brassica rapa subsp rapa Nutrition 0.000 description 2
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- 239000004386 Erythritol Substances 0.000 description 2
- UNXHWFMMPAWVPI-UHFFFAOYSA-N Erythritol Natural products OCC(O)C(O)CO UNXHWFMMPAWVPI-UHFFFAOYSA-N 0.000 description 2
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 102000005640 Myosin Type II Human genes 0.000 description 2
- 108010045128 Myosin Type II Proteins 0.000 description 2
- 101000783356 Naja sputatrix Cytotoxin Proteins 0.000 description 2
- 229930193140 Neomycin Natural products 0.000 description 2
- PVNIIMVLHYAWGP-UHFFFAOYSA-N Niacin Chemical compound OC(=O)C1=CC=CN=C1 PVNIIMVLHYAWGP-UHFFFAOYSA-N 0.000 description 2
- ZCQWOFVYLHDMMC-UHFFFAOYSA-N Oxazole Chemical compound C1=COC=N1 ZCQWOFVYLHDMMC-UHFFFAOYSA-N 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 101710137849 Protein YY1 Proteins 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- MUMGGOZAMZWBJJ-DYKIIFRCSA-N Testostosterone Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 MUMGGOZAMZWBJJ-DYKIIFRCSA-N 0.000 description 2
- 239000005862 Whey Substances 0.000 description 2
- 102000007544 Whey Proteins Human genes 0.000 description 2
- 108010046377 Whey Proteins Proteins 0.000 description 2
- TVXBFESIOXBWNM-UHFFFAOYSA-N Xylitol Natural products OCCC(O)C(O)C(O)CCO TVXBFESIOXBWNM-UHFFFAOYSA-N 0.000 description 2
- 230000001133 acceleration Effects 0.000 description 2
- 239000012190 activator Substances 0.000 description 2
- 230000006978 adaptation Effects 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 239000000783 alginic acid Substances 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- 229960001126 alginic acid Drugs 0.000 description 2
- 150000004781 alginic acids Chemical class 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 239000006227 byproduct Substances 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 239000004202 carbamide Substances 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 239000002340 cardiotoxin Substances 0.000 description 2
- 231100000677 cardiotoxin Toxicity 0.000 description 2
- 239000001913 cellulose Chemical class 0.000 description 2
- 229920002678 cellulose Chemical class 0.000 description 2
- 235000010980 cellulose Nutrition 0.000 description 2
- 235000013351 cheese Nutrition 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- 229960004926 chlorobutanol Drugs 0.000 description 2
- 238000011109 contamination Methods 0.000 description 2
- 230000008602 contraction Effects 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 238000005520 cutting process Methods 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 235000014113 dietary fatty acids Nutrition 0.000 description 2
- 235000013325 dietary fiber Nutrition 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 239000007884 disintegrant Substances 0.000 description 2
- 238000001035 drying Methods 0.000 description 2
- 235000013399 edible fruits Nutrition 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- UNXHWFMMPAWVPI-ZXZARUISSA-N erythritol Chemical compound OC[C@H](O)[C@H](O)CO UNXHWFMMPAWVPI-ZXZARUISSA-N 0.000 description 2
- 235000019414 erythritol Nutrition 0.000 description 2
- 229940009714 erythritol Drugs 0.000 description 2
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 229930195729 fatty acid Natural products 0.000 description 2
- 238000013230 female C57BL/6J mice Methods 0.000 description 2
- 239000000945 filler Substances 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 235000013373 food additive Nutrition 0.000 description 2
- 239000002778 food additive Substances 0.000 description 2
- 235000013355 food flavoring agent Nutrition 0.000 description 2
- 239000003205 fragrance Substances 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 238000003306 harvesting Methods 0.000 description 2
- 235000013402 health food Nutrition 0.000 description 2
- 230000001744 histochemical effect Effects 0.000 description 2
- ZLTPDFXIESTBQG-UHFFFAOYSA-N isothiazole Chemical compound C=1C=NSC=1 ZLTPDFXIESTBQG-UHFFFAOYSA-N 0.000 description 2
- CTAPFRYPJLPFDF-UHFFFAOYSA-N isoxazole Chemical compound C=1C=NOC=1 CTAPFRYPJLPFDF-UHFFFAOYSA-N 0.000 description 2
- 239000004310 lactic acid Substances 0.000 description 2
- 235000014655 lactic acid Nutrition 0.000 description 2
- 210000002429 large intestine Anatomy 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 235000019359 magnesium stearate Nutrition 0.000 description 2
- 238000013227 male C57BL/6J mice Methods 0.000 description 2
- 235000012054 meals Nutrition 0.000 description 2
- 235000013372 meat Nutrition 0.000 description 2
- HEBKCHPVOIAQTA-UHFFFAOYSA-N meso ribitol Natural products OCC(O)C(O)C(O)CO HEBKCHPVOIAQTA-UHFFFAOYSA-N 0.000 description 2
- 230000003387 muscular Effects 0.000 description 2
- 229960004927 neomycin Drugs 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 235000012149 noodles Nutrition 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- LXCFILQKKLGQFO-UHFFFAOYSA-N p-hydroxybenzoic acid methyl ester Natural products COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 2
- 229950010131 puromycin Drugs 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 235000021067 refined food Nutrition 0.000 description 2
- 239000000849 selective androgen receptor modulator Substances 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 210000000813 small intestine Anatomy 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- GEHJYWRUCIMESM-UHFFFAOYSA-L sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 235000010356 sorbitol Nutrition 0.000 description 2
- 239000000600 sorbitol Substances 0.000 description 2
- 229960002920 sorbitol Drugs 0.000 description 2
- 239000007921 spray Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 229960004793 sucrose Drugs 0.000 description 2
- 229910052717 sulfur Inorganic materials 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 239000002562 thickening agent Substances 0.000 description 2
- 150000003852 triazoles Chemical class 0.000 description 2
- 210000000689 upper leg Anatomy 0.000 description 2
- 235000013311 vegetables Nutrition 0.000 description 2
- 239000000811 xylitol Substances 0.000 description 2
- 235000010447 xylitol Nutrition 0.000 description 2
- HEBKCHPVOIAQTA-SCDXWVJYSA-N xylitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)CO HEBKCHPVOIAQTA-SCDXWVJYSA-N 0.000 description 2
- 229960002675 xylitol Drugs 0.000 description 2
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- DDMOUSALMHHKOS-UHFFFAOYSA-N 1,2-dichloro-1,1,2,2-tetrafluoroethane Chemical compound FC(F)(Cl)C(F)(F)Cl DDMOUSALMHHKOS-UHFFFAOYSA-N 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-N 0.000 description 1
- VZSRBBMJRBPUNF-UHFFFAOYSA-N 2-(2,3-dihydro-1H-inden-2-ylamino)-N-[3-oxo-3-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)propyl]pyrimidine-5-carboxamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C(=O)NCCC(N1CC2=C(CC1)NN=N2)=O VZSRBBMJRBPUNF-UHFFFAOYSA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- XZKIHKMTEMTJQX-UHFFFAOYSA-N 4-Nitrophenyl Phosphate Chemical compound OP(O)(=O)OC1=CC=C([N+]([O-])=O)C=C1 XZKIHKMTEMTJQX-UHFFFAOYSA-N 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- JGRPKOGHYBAVMW-UHFFFAOYSA-N 8-hydroxy-5-quinolinecarboxylic acid Chemical compound C1=CC=C2C(C(=O)O)=CC=C(O)C2=N1 JGRPKOGHYBAVMW-UHFFFAOYSA-N 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 239000012109 Alexa Fluor 568 Substances 0.000 description 1
- 108010011485 Aspartame Proteins 0.000 description 1
- 244000075850 Avena orientalis Species 0.000 description 1
- 235000007319 Avena orientalis Nutrition 0.000 description 1
- 235000016068 Berberis vulgaris Nutrition 0.000 description 1
- 241000335053 Beta vulgaris Species 0.000 description 1
- 241001474374 Blennius Species 0.000 description 1
- 239000005996 Blood meal Substances 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 241000195649 Chlorella <Chlorellales> Species 0.000 description 1
- 244000060011 Cocos nucifera Species 0.000 description 1
- 235000013162 Cocos nucifera Nutrition 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 108010082495 Dietary Plant Proteins Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 239000004278 EU approved seasoning Substances 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 229940124602 FDA-approved drug Drugs 0.000 description 1
- 240000008620 Fagopyrum esculentum Species 0.000 description 1
- 235000009419 Fagopyrum esculentum Nutrition 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 239000004606 Fillers/Extenders Substances 0.000 description 1
- 235000019733 Fish meal Nutrition 0.000 description 1
- 229930091371 Fructose Natural products 0.000 description 1
- 239000005715 Fructose Substances 0.000 description 1
- RFSUNEUAIZKAJO-ARQDHWQXSA-N Fructose Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O RFSUNEUAIZKAJO-ARQDHWQXSA-N 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 241000173371 Garcinia indica Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 108010051696 Growth Hormone Proteins 0.000 description 1
- 240000005979 Hordeum vulgare Species 0.000 description 1
- 235000007340 Hordeum vulgare Nutrition 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 244000017020 Ipomoea batatas Species 0.000 description 1
- 235000002678 Ipomoea batatas Nutrition 0.000 description 1
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 240000006240 Linum usitatissimum Species 0.000 description 1
- 235000004431 Linum usitatissimum Nutrition 0.000 description 1
- UPYKUZBSLRQECL-UKMVMLAPSA-N Lycopene Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1C(=C)CCCC1(C)C)C=CC=C(/C)C=CC2C(=C)CCCC2(C)C UPYKUZBSLRQECL-UKMVMLAPSA-N 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 208000002720 Malnutrition Diseases 0.000 description 1
- GUBGYTABKSRVRQ-PICCSMPSSA-N Maltose Natural products O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@@H](CO)OC(O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-PICCSMPSSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 241000237502 Ostreidae Species 0.000 description 1
- 108091012461 PHD finger protein 20 Proteins 0.000 description 1
- 108010067035 Pancrelipase Proteins 0.000 description 1
- 229920002230 Pectic acid Polymers 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 235000010627 Phaseolus vulgaris Nutrition 0.000 description 1
- 244000046052 Phaseolus vulgaris Species 0.000 description 1
- BUGBHKTXTAQXES-UHFFFAOYSA-N Selenium Chemical compound [Se] BUGBHKTXTAQXES-UHFFFAOYSA-N 0.000 description 1
- 244000000231 Sesamum indicum Species 0.000 description 1
- 235000003434 Sesamum indicum Nutrition 0.000 description 1
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 244000061456 Solanum tuberosum Species 0.000 description 1
- 235000002595 Solanum tuberosum Nutrition 0.000 description 1
- 102100038803 Somatotropin Human genes 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 244000228451 Stevia rebaudiana Species 0.000 description 1
- 244000269722 Thea sinensis Species 0.000 description 1
- 101001023030 Toxoplasma gondii Myosin-D Proteins 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 241000209140 Triticum Species 0.000 description 1
- 235000021307 Triticum Nutrition 0.000 description 1
- 206010047486 Virilism Diseases 0.000 description 1
- 240000008042 Zea mays Species 0.000 description 1
- 235000005824 Zea mays ssp. parviglumis Nutrition 0.000 description 1
- 235000002017 Zea mays subsp mays Nutrition 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 229940023476 agar Drugs 0.000 description 1
- 235000010419 agar Nutrition 0.000 description 1
- 235000013334 alcoholic beverage Nutrition 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 235000005550 amino acid supplement Nutrition 0.000 description 1
- 239000003263 anabolic agent Substances 0.000 description 1
- 229940070021 anabolic steroids Drugs 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 235000021405 artificial diet Nutrition 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000000605 aspartame Substances 0.000 description 1
- 235000010357 aspartame Nutrition 0.000 description 1
- IAOZJIPTCAWIRG-QWRGUYRKSA-N aspartame Chemical compound OC(=O)C[C@H](N)C(=O)N[C@H](C(=O)OC)CC1=CC=CC=C1 IAOZJIPTCAWIRG-QWRGUYRKSA-N 0.000 description 1
- 229960003438 aspartame Drugs 0.000 description 1
- 238000011888 autopsy Methods 0.000 description 1
- 239000000022 bacteriostatic agent Substances 0.000 description 1
- 235000015278 beef Nutrition 0.000 description 1
- 239000000440 bentonite Substances 0.000 description 1
- 229910000278 bentonite Inorganic materials 0.000 description 1
- SVPXDRXYRYOSEX-UHFFFAOYSA-N bentoquatam Chemical compound O.O=[Si]=O.O=[Al]O[Al]=O SVPXDRXYRYOSEX-UHFFFAOYSA-N 0.000 description 1
- 229960000686 benzalkonium chloride Drugs 0.000 description 1
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 1
- DHCLVCXQIBBOPH-UHFFFAOYSA-N beta-glycerol phosphate Natural products OCC(CO)OP(O)(O)=O DHCLVCXQIBBOPH-UHFFFAOYSA-N 0.000 description 1
- GHRQXJHBXKYCLZ-UHFFFAOYSA-L beta-glycerolphosphate Chemical compound [Na+].[Na+].CC(CO)OOP([O-])([O-])=O GHRQXJHBXKYCLZ-UHFFFAOYSA-L 0.000 description 1
- GUBGYTABKSRVRQ-QUYVBRFLSA-N beta-maltose Chemical compound OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-QUYVBRFLSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 238000004159 blood analysis Methods 0.000 description 1
- 230000037182 bone density Effects 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 235000008429 bread Nutrition 0.000 description 1
- 238000009395 breeding Methods 0.000 description 1
- 230000001488 breeding effect Effects 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 229960003563 calcium carbonate Drugs 0.000 description 1
- 229910000019 calcium carbonate Inorganic materials 0.000 description 1
- 244000309466 calf Species 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 235000013574 canned fruits Nutrition 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229960004424 carbon dioxide Drugs 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000005473 carotenes Nutrition 0.000 description 1
- 150000001746 carotenes Chemical class 0.000 description 1
- 239000005018 casein Substances 0.000 description 1
- BECPQYXYKAMYBN-UHFFFAOYSA-N casein, tech. Chemical compound NCCCCC(C(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(CC(C)C)N=C(O)C(CCC(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(C(C)O)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(COP(O)(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(N)CC1=CC=CC=C1 BECPQYXYKAMYBN-UHFFFAOYSA-N 0.000 description 1
- 235000021240 caseins Nutrition 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 239000008004 cell lysis buffer Substances 0.000 description 1
- SCKYRAXSEDYPSA-UHFFFAOYSA-N ciclopirox Chemical compound ON1C(=O)C=C(C)C=C1C1CCCCC1 SCKYRAXSEDYPSA-UHFFFAOYSA-N 0.000 description 1
- 229960003749 ciclopirox Drugs 0.000 description 1
- 239000010941 cobalt Substances 0.000 description 1
- 229910017052 cobalt Inorganic materials 0.000 description 1
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000010835 comparative analysis Methods 0.000 description 1
- 235000009508 confectionery Nutrition 0.000 description 1
- 235000014510 cooky Nutrition 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 235000005822 corn Nutrition 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 229940099112 cornstarch Drugs 0.000 description 1
- 235000013365 dairy product Nutrition 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- FMGSKLZLMKYGDP-USOAJAOKSA-N dehydroepiandrosterone Chemical compound C1[C@@H](O)CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CC=C21 FMGSKLZLMKYGDP-USOAJAOKSA-N 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- UMNKXPULIDJLSU-UHFFFAOYSA-N dichlorofluoromethane Chemical compound FC(Cl)Cl UMNKXPULIDJLSU-UHFFFAOYSA-N 0.000 description 1
- 229940099364 dichlorofluoromethane Drugs 0.000 description 1
- 229940087091 dichlorotetrafluoroethane Drugs 0.000 description 1
- 235000019621 digestibility Nutrition 0.000 description 1
- 229940079920 digestives acid preparations Drugs 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 238000002224 dissection Methods 0.000 description 1
- 239000008298 dragée Substances 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 235000006694 eating habits Nutrition 0.000 description 1
- 239000008157 edible vegetable oil Substances 0.000 description 1
- 238000001378 electrochemiluminescence detection Methods 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 210000003746 feather Anatomy 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 238000011049 filling Methods 0.000 description 1
- 239000004467 fishmeal Substances 0.000 description 1
- 235000004426 flaxseed Nutrition 0.000 description 1
- 239000004088 foaming agent Substances 0.000 description 1
- 235000011194 food seasoning agent Nutrition 0.000 description 1
- 235000013611 frozen food Nutrition 0.000 description 1
- 235000011389 fruit/vegetable juice Nutrition 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 239000003349 gelling agent Substances 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000008202 granule composition Substances 0.000 description 1
- 238000000227 grinding Methods 0.000 description 1
- 239000000122 growth hormone Substances 0.000 description 1
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 229960002725 isoflurane Drugs 0.000 description 1
- 239000000644 isotonic solution Substances 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 239000000865 liniment Substances 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 229940057995 liquid paraffin Drugs 0.000 description 1
- 210000005228 liver tissue Anatomy 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 238000011866 long-term treatment Methods 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000000395 magnesium oxide Substances 0.000 description 1
- CPLXHLVBOLITMK-UHFFFAOYSA-N magnesium oxide Inorganic materials [Mg]=O CPLXHLVBOLITMK-UHFFFAOYSA-N 0.000 description 1
- AXZKOIWUVFPNLO-UHFFFAOYSA-N magnesium;oxygen(2-) Chemical compound [O-2].[Mg+2] AXZKOIWUVFPNLO-UHFFFAOYSA-N 0.000 description 1
- 239000002075 main ingredient Substances 0.000 description 1
- 230000001071 malnutrition Effects 0.000 description 1
- 235000000824 malnutrition Nutrition 0.000 description 1
- 235000010449 maltitol Nutrition 0.000 description 1
- 239000000845 maltitol Substances 0.000 description 1
- VQHSOMBJVWLPSR-WUJBLJFYSA-N maltitol Chemical compound OC[C@H](O)[C@@H](O)[C@@H]([C@H](O)CO)O[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O VQHSOMBJVWLPSR-WUJBLJFYSA-N 0.000 description 1
- 229940035436 maltitol Drugs 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 229960001855 mannitol Drugs 0.000 description 1
- 235000013310 margarine Nutrition 0.000 description 1
- 239000003264 margarine Substances 0.000 description 1
- 231100000794 masculinization Toxicity 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 208000030159 metabolic disease Diseases 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 239000012120 mounting media Substances 0.000 description 1
- 230000004118 muscle contraction Effects 0.000 description 1
- 230000009753 muscle formation Effects 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 235000001968 nicotinic acid Nutrition 0.000 description 1
- 239000011664 nicotinic acid Substances 0.000 description 1
- 229960003512 nicotinic acid Drugs 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 235000016709 nutrition Nutrition 0.000 description 1
- 208000015380 nutritional deficiency disease Diseases 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 235000020636 oyster Nutrition 0.000 description 1
- 239000003002 pH adjusting agent Substances 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 235000015927 pasta Nutrition 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- LCLHHZYHLXDRQG-ZNKJPWOQSA-N pectic acid Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)O[C@H](C(O)=O)[C@@H]1OC1[C@H](O)[C@@H](O)[C@@H](OC2[C@@H]([C@@H](O)[C@@H](O)[C@H](O2)C(O)=O)O)[C@@H](C(O)=O)O1 LCLHHZYHLXDRQG-ZNKJPWOQSA-N 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 239000008389 polyethoxylated castor oil Substances 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000010318 polygalacturonic acid Substances 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Chemical class 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 229920001592 potato starch Polymers 0.000 description 1
- 229940116317 potato starch Drugs 0.000 description 1
- 235000012015 potatoes Nutrition 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 239000006041 probiotic Substances 0.000 description 1
- 235000018291 probiotics Nutrition 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004405 propyl p-hydroxybenzoate Substances 0.000 description 1
- 229960003415 propylparaben Drugs 0.000 description 1
- 208000026455 prostate symptom Diseases 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 238000002731 protein assay Methods 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 238000010298 pulverizing process Methods 0.000 description 1
- 210000003314 quadriceps muscle Anatomy 0.000 description 1
- 238000011158 quantitative evaluation Methods 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 230000035484 reaction time Effects 0.000 description 1
- HELXLJCILKEWJH-NCGAPWICSA-N rebaudioside A Chemical compound O([C@H]1[C@H](O)[C@@H](CO)O[C@H]([C@@H]1O[C@H]1[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O1)O)O[C@]12C(=C)C[C@@]3(C1)CC[C@@H]1[C@@](C)(CCC[C@]1([C@@H]3CC2)C)C(=O)O[C@H]1[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O1)O)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O HELXLJCILKEWJH-NCGAPWICSA-N 0.000 description 1
- 238000007670 refining Methods 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- 229940100486 rice starch Drugs 0.000 description 1
- 235000019204 saccharin Nutrition 0.000 description 1
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 1
- 229940081974 saccharin Drugs 0.000 description 1
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 235000015067 sauces Nutrition 0.000 description 1
- 235000013580 sausages Nutrition 0.000 description 1
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical compound O1C(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(O)C2O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC2C(O)C(O)C1OC2CO HFHDHCJBZVLPGP-UHFFFAOYSA-N 0.000 description 1
- 229910052711 selenium Inorganic materials 0.000 description 1
- 239000011669 selenium Substances 0.000 description 1
- 239000003352 sequestering agent Substances 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 239000004460 silage Substances 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 229940079827 sodium hydrogen sulfite Drugs 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- CMZUMMUJMWNLFH-UHFFFAOYSA-N sodium metavanadate Chemical compound [Na+].[O-][V](=O)=O CMZUMMUJMWNLFH-UHFFFAOYSA-N 0.000 description 1
- 229940001482 sodium sulfite Drugs 0.000 description 1
- 235000010265 sodium sulphite Nutrition 0.000 description 1
- 239000012439 solid excipient Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 235000013555 soy sauce Nutrition 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 239000008227 sterile water for injection Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000010902 straw Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000005846 sugar alcohols Chemical class 0.000 description 1
- 239000007940 sugar coated tablet Substances 0.000 description 1
- 229960002673 sulfacetamide Drugs 0.000 description 1
- SKIVFJLNDNKQPD-UHFFFAOYSA-N sulfacetamide Chemical compound CC(=O)NS(=O)(=O)C1=CC=C(N)C=C1 SKIVFJLNDNKQPD-UHFFFAOYSA-N 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 210000002435 tendon Anatomy 0.000 description 1
- 229960003604 testosterone Drugs 0.000 description 1
- 239000000892 thaumatin Substances 0.000 description 1
- 235000010436 thaumatin Nutrition 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 210000000115 thoracic cavity Anatomy 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 231100000820 toxicity test Toxicity 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- CYRMSUTZVYGINF-UHFFFAOYSA-N trichlorofluoromethane Chemical compound FC(Cl)(Cl)Cl CYRMSUTZVYGINF-UHFFFAOYSA-N 0.000 description 1
- 229940029284 trichlorofluoromethane Drugs 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 229960004418 trolamine Drugs 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 235000019155 vitamin A Nutrition 0.000 description 1
- 239000011719 vitamin A Substances 0.000 description 1
- NCYCYZXNIZJOKI-UHFFFAOYSA-N vitamin A aldehyde Natural products O=CC=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C NCYCYZXNIZJOKI-UHFFFAOYSA-N 0.000 description 1
- 235000019156 vitamin B Nutrition 0.000 description 1
- 239000011720 vitamin B Substances 0.000 description 1
- 229940046001 vitamin b complex Drugs 0.000 description 1
- 239000000341 volatile oil Substances 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 208000016261 weight loss Diseases 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
- 235000015099 wheat brans Nutrition 0.000 description 1
- 229940100445 wheat starch Drugs 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 229910000166 zirconium phosphate Inorganic materials 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/63—Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/63—Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
- A61K31/635—Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
Definitions
- the present disclosure relates to a composition for improving, treating or preventing muscular disorders including sulfonamide-based compounds.
- Sarcopenia is a disease in which the muscle mass is reduced or the muscle strength decreases due to diseases, aging, and the like.
- the muscle mass gradually decreases after the age of 40's, and it is estimated that a decrease of 8% in the muscle mass occurs every 10 years until the age of 70's, and thereafter, a more rapid decrease in the muscle mass occurs, and it is known that the decrease in the muscle mass may occur up to 15% every 10 years.
- Senile sarcopenia causes not only direct muscle strength reduction to increase the risk of death due to reduction and disability of various body functions, but also a decrease in metabolism and a decrease in immunity to increase the prevalence of metabolic diseases such as hypertension, diabetes, arthritis, obesity, and cancer.
- the senile sarcopenia is a disease that occurs in 40% of the elderly over 80 years of age and is expected to increase social and economic burdens in the aging era.
- PHD finger protein 20 is a protein also known as glioma-expressed antigen 2 (GLEA2). Recently, it has been found that PHF 20, as a transcription factor, acts on muscle damage and sarcopenia by regulating a sub-target protein YY1. It has been reported that the expression of PHF20 increases and then decreases during muscle cell differentiation using myoblasts (C2C12), and this phenomenon is regulated together with the expression of a muscle differentiation inhibitory protein YY1.
- sarcopenia there are three major treatment methods for sarcopenia.
- the first is exercise. It has been reported that the exercise increases the protein synthesis ability of skeletal muscle in the short term, and increases muscle strength or mobility in the elderly. However, the exercise is not suitable for long-term treatment.
- testosterone or anabolic steroids can be used as drug treatment, but induces masculinization in women and causes side effects such as prostate symptoms in men.
- Other approved prescriptions include dehydroepiandrosterone (DHEA) and growth hormone, which have been reported in studies that can be used as treatment methods at sites containing selective androgen receptor modulators (SARMs).
- DHEA dehydroepiandrosterone
- SARMs selective androgen receptor modulators
- diet therapy is known as a treatment method, according to nutritional assessments, malnutrition, and modern eating habits are inadequate to maintain adequate total body mass. However, the reality is that there is no fundamental therapeutic agent or enhancer for sarcopenia.
- the present inventors of the present disclosure have screen materials that may restore or alleviate the inhibition of muscle differentiation under conditions that muscle differentiation in myoblasts is inhibited by PHF20 overexpression, by using a screening system constructed using PHF20/YY1. Accordingly, the present inventors confirmed that the selected compounds could be used to inhibit muscle reduction and promote muscle differentiation, and completed the present disclosure.
- a pharmaceutical composition for prevention or treatment that may help treat, relieve, alleviate or prevent diseases, including a sulfonamide-based compound represented by Chemical Formula 1 below or a salt thereof; or a food composition; a quasi-drug composition; a feed composition; or a composition for feed additives capable of preventing, improving, relieving or alleviating desired symptoms or assisting in the desired symptoms.
- an objective of the present disclosure is to provide a pharmaceutical composition for preventing or treating muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 below or a pharmaceutically acceptable salt thereof.
- another objective of the present disclosure is to provide a food composition for preventing or improving muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 below or a food acceptable salt thereof.
- yet another objective of the present disclosure is to provide a feed composition for preventing or improving muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 below or a salt thereof.
- prevention refers to all actions that inhibit the development of muscular disorders by administering the composition, or inhibit or delay the severity of muscular disorders.
- treatment refers to all actions that improve or beneficially change the symptoms of muscular disorders by administration of the composition.
- “capable of assisting” means to inhibit or delay symptoms of muscular disorders by administering the composition of the present disclosure, or to help or be able to help in improving the symptoms caused by muscular disorders. It may be used as an adjuvant to enhance an effect of medicines for treatment, and has meaning as an adjuvant as a health functional food or functional food.
- the composition for preventing, improving or treating muscular disorders including the sulfonamide-based compounds or salts thereof can prevent inhibition of differentiation of myoblasts by regulating the expression of PHF20 and YY1. Accordingly, since the composition may prevent or alleviate muscle loss, or promote muscle regeneration to improve muscle functions and muscle mass, the composition can be effectively used for therapeutic agents, foods, or feeds for prevention, improvement or treatment of muscle disorders, and improving muscle functions or muscle mass.
- the present disclosure provides a use for preventing, improving or treating muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 or a salt thereof; and a composition for preventing, improving or treating muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 or a salt thereof.
- the hetero element of the hetero aryl may be one or more selected from S, N, and O, one or more hetero elements may be included, and one or more hetero elements different from each other may be included.
- the aryl or heteroaryl may be selected from the group consisting of pyrrole, pyrazole, triazole, oxazole, furan, isoxazole, isothiazole, imidazole, diazole, thiadiazole, phenyl, pyridine, pyrimidine, triazine, oxazine, and thiazine.
- the aryl or heteroaryl may be thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine.
- the substituent of R 1 may be at least one substituent selected from C 1-3 alkyl, C 1-3 alkoxy, and phenyl. In a preferred embodiment, the substituent of R 1 may be at least one selected from the group consisting of methyl, ethyl, methoxy, ethoxy and phenyl. One substituent of R 1 may be included, and one or more substituents identical to or different from each other may be included.
- the compound of Chemical Formula 1 may be sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide or sulfadiazine.
- the salt of the compound of the present disclosure includes all kinds of salts commonly accepted in the art of the present disclosure, and may be, for example, a sodium salt.
- the sulfasalazine may be a compound represented by Chemical Formula 2 below.
- the sulfamethazine may be a compound represented by Chemical Formula 3 below.
- the sulfathiazole may be a compound represented by Chemical Formula 4 below.
- the sulfapyridine may be a compound represented by Chemical Formula 5 below.
- the sulfaphenazole may be a compound represented by Chemical Formula 6 below.
- the sulfameter may be a compound represented by Chemical Formula 7 below.
- the sulfamethizole may be a compound represented by Chemical Formula 8 below.
- the sulfaguanidine may be a compound represented by Chemical Formula 9 below.
- the sulfacetamide may exist in the form of a sodium salt, and more specifically, may be a compound represented by Chemical Formula 10 below.
- the sulfadoxin may be a compound represented by Chemical Formula 11 below.
- the sulfadimethoxine may be a compound represented by Chemical Formula 12 below.
- the sulfanilamide may be a compound represented by Chemical Formula 13 below.
- the sulfadiazine may be a compound represented by Chemical Formula 14 below.
- the muscular disorder means a disease or symptom that causes a reduced muscle function; a decrease in muscle mass; muscle wasting; or muscle degeneration, or worsens these symptoms.
- the term 'muscle' collectively refers to tendons and muscles.
- the ⁇ muscle function' refers to the ability to exert power by contraction of muscles, and includes muscular strength which is the ability of the muscles to exhibit maximum contraction to overcome resistance, muscular endurance which is the ability to exhibit how long or how many times muscles may repeat contraction and relaxation at a given weight, and quickness which is the ability to exert strong power in a short time.
- the term ⁇ improvement of muscle function' refers to improving the muscle function better by increasing the muscle mass, promoting regeneration of damaged muscles, or improving regeneration.
- the ⁇ promoting the muscle regeneration' means shortening the regeneration or recovery time of damaged muscles, increasing muscle mass by activating the muscles, or lowering the degree of muscle mass loss.
- the muscle wasting and degeneration are caused by genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle, but is not limited to a type of muscle.
- the muscular disorder may be one selected from the group consisting of sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia.
- the present disclosure provides a pharmaceutical composition for preventing or treating muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 or a salt thereof; a food composition for preventing or treating muscular disorders; and a feed composition for preventing or treating muscular disorders:
- the hetero element of the hetero aryl may be one or more selected from S, N, and O, one or more hetero elements may be included, and one or more hetero elements different from each other may be included.
- the aryl or heteroaryl may be selected from the group consisting of pyrrole, pyrazole, triazole, oxazole, furan, isoxazole, isothiazole, imidazole, diazole, thiadiazole, phenyl, pyridine, pyrimidine, triazine, oxazine, and thiazine.
- the aryl or heteroaryl may be thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine.
- the substituent of R 1 may be at least one substituent selected from C 1-3 alkyl, C 1-3 alkoxy, and phenyl. In a preferred embodiment, the substituent of R 1 may be at least one selected from the group consisting of methyl, ethyl, methoxy, ethoxy and phenyl. One substituent of R 1 may be included, and one or more substituents identical to or different from each other may be included.
- the compound of Chemical Formula 1 may be sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide or sulfadiazine.
- the salt of the compound of the present disclosure includes all kinds of salts commonly accepted in the art of the present disclosure, and may be, for example, a sodium salt.
- the compound of Chemical Formula 1 of the present disclosure acts on PHD20/YY1 to suppress the inhibition of the formation of myoblasts, thereby inhibiting a decrease in muscles in muscle tissues or promoting muscle formation.
- the sulfamethazine of the present disclosure acts on PHD20/YY1 to suppress the inhibition of the formation of myoblasts, thereby inhibiting muscle damage and promoting regeneration, and as a result, it was confirmed in vivo that the balance ability and grip strength recovery of Rota Rod were superior to those of muscle-damaged mice.
- the composition for preventing or treating the muscular disorders of the present disclosure is the pharmaceutical composition
- the composition may be used for preventing or treating muscular disorders caused by muscle wasting or degeneration.
- the muscle wasting and degeneration occur due to genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle.
- Examples of related disorders may include sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia, but are not limited thereto.
- the pharmaceutical composition of the present disclosure may include a pharmaceutically acceptable carrier.
- the pharmaceutically acceptable carrier may further include, for example, a carrier for oral administration or a carrier for parenteral administration.
- the carrier for oral administration may include lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like.
- the carrier for parenteral administration may include water, suitable oil, saline, aqueous glucose, glycol, and the like.
- a stabilizer and a preservative may be further included.
- a suitable stabilizer includes antioxidants such as sodium hydrogen sulfite, sodium sulfite or ascorbic acid.
- a suitable preservative includes benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol.
- the pharmaceutical composition of the present disclosure may be administered to mammals including humans even by any method.
- the pharmaceutical composition may be administered orally or parenterally, and the parenteral administration method is not limited thereto, but may include intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, intestinal, topical, sublingual or rectal administration.
- the pharmaceutical composition of the present disclosure may be formulated as formulations for oral administration or parenteral administration according to the route of administration as described above.
- the formulation may be prepared by using one or more buffers (e.g., saline or PBS), antioxidants, bacteriostatic agents, chelating agents (e.g., EDTA or glutathione), fillers, extenders, binders, adjuvants (e.g., aluminum hydroxide), suspending agents, thickening agents, wetting agents, disintegrants or surfactants, and diluents or excipients.
- buffers e.g., saline or PBS
- antioxidants e.g., bacteriostatic agents, chelating agents (e.g., EDTA or glutathione)
- fillers e.g., extenders, binders, adjuvants (e.g., aluminum hydroxide), suspending agents, thickening agents, wetting agents, disintegrants or surfactants, and
- Solid preparations for oral administration include tablets, pills, powders, granules, liquids, gels, syrups, slurries, suspensions or capsules. These solid preparations may be prepared by mixing at least one excipient, for example, starch (including corn starch, wheat starch, rice starch, potato starch, etc.), calcium carbonate, sucrose, lactose, dextrose, sorbitol, mannitol, xylitol, erythritol maltitol, cellulose, methyl cellulose, sodium carboxymethyl cellulose and hydroxypropyl methyl cellulose or gelatin with the pharmaceutical composition of the present disclosure.
- tablets or sugar-coated tablets may be obtained by mixing an active ingredient with a solid excipient, pulverizing the mixture, adding a suitable adjuvant, and then processing the mixture into a granule mixture.
- Lubricants such as magnesium stearate and talc is also used in addition to simple excipients.
- Liquid preparations for oral administration may correspond to suspensions, oral liquids, emulsions, syrups, or the like, and may include various excipients, for example, a wetting agent, a sweetener, an aromatic agent, and a preserving agent, in addition to water or liquid paraffin which is a commonly used simple diluent.
- cross-linked polyvinylpyrrolidone, agar, alginic acid or sodium alginate may be added as a disintegrant, and an anti-coagulant, a lubricant, a wetting agent, a fragrance, an emulsifier, a preservative, and the like may be additionally included.
- the pharmaceutical composition of the present disclosure when administered parenterally, may be formulated according to a method known in the art in the form of injections, transdermal agents, and nasal inhalers together with suitable parenteral carriers.
- the injections need to be sterilized and protected from contamination of microorganisms such as bacteria and fungi.
- suitable carriers for injections are not limited thereto, but may be solvents or dispersion media containing water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols), mixtures thereof and/or vegetable oils.
- a Hanks' solution a Ringer's solution, a phosphate buffered saline (PBS) containing triethanol amine or sterile water for injection, and an isotonic solution such as 10% ethanol, 40% propylene glycol and 5% dextrose may be used.
- PBS phosphate buffered saline
- an isotonic solution such as 10% ethanol, 40% propylene glycol and 5% dextrose
- various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid, and thimerosal may be further included.
- most of the injections may further include an isotonic agent, such as sugar or sodium chloride.
- the transdermal agents are included in the form of ointments, creams, lotions, gels, external liquids, pastas, liniments, and aerosols.
- the ⁇ transdermal administration' means that an effective dose of the active ingredient contained in the pharmaceutical composition is delivered into the skin by topically applying the pharmaceutical composition to the skin.
- the compound used according to the present disclosure may be conveniently delivered in the form of an aerosol spray from a pressurized pack or a nebulizer by using a suitable propellant, for example, dichlorofluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, and carbon dioxide, or other suitable gases.
- a dosage unit may be determined by providing a valve that delivers a metered amount.
- gelatin capsules and cartridges used in an inhaler or insufflator may be formulated to contain a powder mixture of a compound and a suitable powder base such as lactose or starch.
- the pharmaceutical composition for preventing or treating the muscular disorders of the present disclosure may provide preferred preventive or therapeutic effects on muscular disorders when including an effective dose of the compound of Chemical Formula 1.
- a method for treating muscular disorders including administering the compound represented by Chemical Formula 1 or a pharmaceutically acceptable salt thereof to a subject in a therapeutically effective dose.
- the treatment method may further include identifying a subject in need of prevention or treatment of the muscular disorders before the administering step.
- the ⁇ therapeutically effective dose' or ⁇ effective dose' refers to an amount that exhibits a greater response than that of a negative control group, and preferably refers to an amount sufficient to improve muscle function.
- the pharmaceutical composition of the present disclosure may contain 0.01 to 99.99% of the compound of Chemical Formula 1 or the pharmaceutically acceptable salt thereof, and the remaining amount may account for the pharmaceutically acceptable carrier.
- the effective dose of the compound of Chemical Formula 1 or the pharmaceutically acceptable salt thereof included in the pharmaceutical composition of the present disclosure may vary depending on a form in which the composition is commercialized.
- the total effective dose of the pharmaceutical composition of the present disclosure may be administered to a subject in a single dose, or may be administered to the subject in a multiple dose for a long period of time according to a fractionated treatment protocol.
- the content of the active ingredient may vary depending on the severity of disease.
- an effective dose to the subject may be determined by considering various factors such as the age, weight, health condition, sex, severity of disease, diet and excretion rate of the subject as well as the route of administration of the pharmaceutical composition and the number of treatments. Considering these points, those skilled in the art may determine an appropriate effective dose of the compound of Chemical Formula 1 or the pharmaceutically acceptable salt thereof according to a specific use for preventing or treating muscular disorders.
- the pharmaceutical composition according to the present disclosure is not particularly limited to the formulation, the administration route, and the administration method thereof, as long as the effects of the present disclosure are shown.
- the “subject” may refer to mammals such as human or non-human primates, mice, dogs, cats, horses, and cows, but is not limited thereto.
- the pharmaceutical composition for preventing or treating the muscular disorders of the present disclosure may also be provided in a formulation for external use including the compound of Chemical Formula 1 or the pharmaceutically acceptable salt thereof as an active ingredient.
- the pharmaceutical composition of the present disclosure may further contain adjuvants commonly used in the field of dermatology, such as any other ingredients commonly used in external skin preparations, such as fatty substances, organic solvents, solubilizers, concentrate and gelling agents, emollients, antioxidants, suspending agents, stabilizers, foaming agents, fragrances, surfactants, water, ionic emulsifiers, nonionic emulsifiers, fillers, sequestering agents, chelating agents, preservatives, vitamins, blockers, wetting agents, essential oils, dyes, pigments, hydrophilic activators, lipophilic activators or lipid vesicles.
- the ingredients may be introduced in an amount generally used in the field of dermatology.
- the external skin preparation is not limited thereto, but may be a formulation such as ointments, patches, gels, creams, or sprays.
- a composition for preventing or improving muscular disorders of the present disclosure may be a food composition for preventing or improving muscular disorders including a compound of Chemical Formula 1 or a food acceptable salt thereof.
- the composition for preventing or treating the muscular disorders of the present disclosure is the food composition
- the composition may be used for preventing or treating muscular disorders caused by muscle wasting or degeneration.
- the muscle wasting and degeneration occur due to genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle.
- Examples of related disorders may include sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia, but are not limited thereto.
- the food composition means including all forms such as functional foods, nutritional supplements, health foods, food additives, or feeds, and is taken for animals including humans or livestock.
- the type of food composition may be prepared in various forms according to a general method known in the art.
- General foods are not limited thereto, but may be prepared by adding the compound of Chemical Formula 1 of the present disclosure or the food acceptable salt thereof to beverages (including alcoholic beverages), fruits and processed foods thereof (e.g., canned fruit, bottled food, jam, and marmalade), fish, meat and processed foods thereof (e.g. ham, sausage, and corned beef), bread and noodles (e.g. udon, buckwheat noodles, ramen, spaghetti, and macaroni), fruit juice, various drinks, cookies, sweets, dairy products (e.g.
- the nutritional supplements are not limited thereto, but may be prepared by adding the compound of Chemical Formula 1 of the present disclosure or the food acceptable salt thereof to capsules, tablets, pills, etc.
- the health functional food is not limited thereto, but for example, may be taken by liquefying, granulating, encapsulating, and powdering so as to be drunken (health beverages) by preparing the compound of Chemical Formula 1 of the present disclosure or the food acceptable salt thereof in the form of tea, juice, and drinks.
- the health functional food may be prepared and used in the form of powders or concentrates.
- the health functional food may be prepared in the form of a composition by mixing the compound of Chemical Formula 1 of the present disclosure or the food acceptable salt thereof with known active ingredients known to have an effect of preventing or improving muscular disorders.
- the health drink composition may contain various flavoring agents or natural carbohydrates as an additional ingredient, like a general drink.
- the natural carbohydrates may be monosaccharides such as glucose and fructose; disaccharides such as maltose and sucrose; polysaccharides such as dextrin and cyclodextrin; and sugar alcohols such as xylitol, sorbitol, and erythritol.
- sweetening agent natural sweetening agents such as thaumatin and stevia extracts; synthetic sweetening agents such as saccharin and aspartame may be used.
- a ratio of the natural carbohydrates may be generally about 0.01 to 0.04 g, preferably about 0.02 to 0.03 g per 100 mL of the composition of the present disclosure.
- the compound represented by Chemical Formula 1 of the present disclosure or the food acceptable salt thereof may be included as an active ingredient in the food composition for preventing or improving the muscular disorders, but the amount is not particularly limited to an amount effective to achieve an action for preventing or improving muscular disorders, but is preferably 0.01 to 100 wt% based on the total weight of the total composition.
- the food composition of the present disclosure may be prepared by mixing the compound represented by Chemical Formula 1 or the food acceptable salt thereof with other active ingredients known to be effective in the composition for preventing or improving the muscular disorders.
- the food composition may contain various nutrients, vitamins, electrolytes, flavoring agents, coloring agents, pectic acid and salts thereof, alginic acid and salts thereof, organic acid, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohol, carbonic acid agents, or the like.
- the health food of the present disclosure may contain pulp for preparing natural fruit juice, fruit juice beverage, or vegetable beverage. These ingredients may be used independently or in combination. Although the ratio of these additives is not very important, generally, the ratio thereof is selected in a range of 0.01 to 0.1 part by weight per 100 parts by weight of the composition of the present disclosure.
- composition for preventing or improving muscular disorders of the present disclosure may be a feed composition or a feed additive composition for preventing or improving muscular disorders including the compound of Chemical Formula 1.
- the term 'feed' may mean any natural or artificial diet, one-meal diet, or ingredients of one-meal diet to be eaten, ingested, and digested by animals or suitable therefor.
- the feed including the composition for preventing or improving the muscular disorders according to the present disclosure as an active ingredient may be prepared with various types of feed known in the art, and preferably, may include concentrated feeds, roughage and/or special feeds, but is not limited thereto.
- ⁇ feed additives' includes substances added to the feeds for the purpose of various effects, such as nutrient supplementation and weight loss prevention, improvement of digestibility of fiber in feed, oil quality improvement, reproduction disorder prevention and conception rate improvement, and summer high temperature stress prevention.
- the feed additives composition of the present disclosure correspond to supplementary feeds under the Feed Management Act, and may further include mineral preparations such as sodium bicarbonate, bentonite, magnesium oxide, and complex minerals, mineral preparations which are trace minerals such as zinc, copper, cobalt, and selenium, vitamins such as carotene, vitamins A, D, E, nicotinic acid, and vitamin B complex, protected amino acid supplements such as methionine and lysine, protected fatty acid preparations such as fatty acid calcium salts, active bacteria such as probiotics (lactic acid bacteria), yeast cultures, and mold fermented products, yeast agents, and the like.
- mineral preparations such as sodium bicarbonate, bentonite, magnesium oxide, and complex minerals
- mineral preparations which are trace minerals such as zinc, copper, cobalt, and selenium such as carotene, vitamins A, D, E, nicotinic acid, and vitamin B complex
- protected amino acid supplements such as methionine and lysine
- the concentrated feeds include seed fruits including grains such as wheat, oats and corn, bran including rice bran, wheat bran, and barley bran as by-products obtained by refining grains, residues such as seed cakes which are by-products obtained from oil extraction of beans, oil, sesame, linseed, and coco palms, and residual starches as a main ingredient of starch residues which are the remainder after removing starch from sweet potatoes and potatoes, fish soluble which is a concentrate of fresh liquids obtained from fish meal, fish residues, and fish, animal feeds such as meat meal, blood meal, feather meal, skim milk powder, dried whey, which is obtained by drying whey as the balance when preparing cheese from milk and casein from skim milk, yeast, chlorella, and seaweed, but are not limited thereto.
- seed fruits including grains such as wheat, oats and corn, bran including rice bran, wheat bran, and barley bran as by-products obtained by refining grains, residues such as seed cakes which
- the roughage includes raw grass feed such as wild grass, grass, and green cutting, root vegetables such as turnips for feed, beets for feed, and Lutherbearer as a type of turnip, silage which is stored feed made by filling a silo with green grass, green crops, and grains, and fermenting the silo with lactic acid, dried hay made by cutting and drying wild grass and grass, straw of breeding crops, and leaves of leguminous plants, but is not limited thereto.
- raw grass feed such as wild grass, grass, and green cutting
- root vegetables such as turnips for feed, beets for feed, and Lutherbearer as a type of turnip
- silage which is stored feed made by filling a silo with green grass, green crops, and grains, and fermenting the silo with lactic acid, dried hay made by cutting and drying wild grass and grass, straw of breeding crops, and leaves of leguminous plants, but is not limited thereto.
- the special feed includes mineral feeds such as oyster shells and rock salt, urea feeds such as urea or its derivative, diureideisobutane, feed additives which are substances supplemented with ingredients that tend to be insufficient when only natural feed raw materials are mixed, or added in a small amount to the formulated feed to improve the storage of feeds, and dietary supplements, but is not limited thereto.
- mineral feeds such as oyster shells and rock salt
- urea feeds such as urea or its derivative, diureideisobutane
- feed additives which are substances supplemented with ingredients that tend to be insufficient when only natural feed raw materials are mixed, or added in a small amount to the formulated feed to improve the storage of feeds, and dietary supplements, but is not limited thereto.
- the feed additives for preventing or improving the muscular disorders according to the present disclosure may be prepared by adding the compound of Chemical Formula 1 of the present disclosure or the salt thereof in an appropriate effective concentration range according to various feed preparation methods known in the art.
- HTS high throughput screening
- a mouse normal myoblast C2C12 cell line was obtained from ATCC (Manassas, VA, USA). C2C12 cells were subcultured every 2 to 3 days in a 100 mm dish containing a DMEM supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 unit/ml penicillin and 100 ⁇ g/ml streptomycin (Life Technologies, Grand Island, NY, USA). The cells were cultured and grown under 5% CO 2 and 37°C conditions in a humidified state. While used in experiments, the cells were collected at the end of treatment for additional analysis.
- C2C12 myoblasts which were stable cells in which PHF20 was overexpressed, were transformed with a plasmid coexisting with a YY1 promoter and GFP, which inhibited muscle differentiation, to prepare a stable cell line.
- the degree of GFP signal suppression was quantified and measured, and the degree of muscle differentiation was used as a quantitative measurement index ( FIG. 1 ).
- an HTS system capable of being used as a measurement index of a quantitative measurement system of the degree of muscle differentiation
- a stable cell line was selected by treatment with hygromycin, a selection marker for GFP expression, by concentration.
- C2C12 cell lines transformed with PHF20 and YY1-promoter-GFP plasmid were treated with 150 ⁇ g/ml (1 set), 50 ⁇ g/ml (2 set) or 250 ⁇ g/ml (3 set) of hygromycin together with 1 mg/ml of neomycin and 2 ⁇ g/ml of puromycin, respectively, to select only perfectly transformed cell lines.
- the medium was replaced once every two days, and neomycin, puromycin, and hygromycin were treated at each concentration, and proceeded for about a month.
- C2C12-PHF20/YY1-promoter-GFP cell lines of three types 150 ⁇ g/ml (1 set), 50 ⁇ g/ml (2 set) or 250 ⁇ g/ml (3 set)) selected separately were dispensed in a 96-well white plate in 0.5 ⁇ 10 5 , cultured for 24 hours, and cells in a 70% confluent state were treated with doxycycline at 0, 50 ng/ml, 250 ng/ml, 500 ng/ml, or 1000 ng/ml, and then cultured for 24 hours.
- C2C12 cells were left on ice, washed twice with cold PBS, an then dissolved at 4°C for 30 minutes in a cell lysis buffer (50 mmol/L Tris-HCl, pH 7.5, 1% (v/v) Nonidet P-40, 250 mmol/L NaCl, 0.1 mmol/L phenyl methyl sulfonyl fluoride, 0.1 mmol/L sodium vanadate, 20 mmol/L ⁇ -glycerol phosphate, 2 mmol/L DTT, 1 mmol/L Leupeptin and 10 mmol/L PNPP).
- a cell lysis buffer 50 mmol/L Tris-HCl, pH 7.5, 1% (v/v) Nonidet P-40, 250 mmol/L NaCl, 0.1 mmol/L phenyl methyl sulfonyl fluoride, 0.1 mmol/L sodium vanadate, 20 mmol/L ⁇ -g
- the cell lysate was centrifuged at 16,000 ⁇ g at 4°C for 20 minutes. The supernatant was collected and used for SDS-PAGE, and the protein content was evaluated by bovine serum albumin protein assay. Proteins were mixed with a sample buffer containing ⁇ -mercamptoethanol and heated at 100°C for 2 minutes. 40 ⁇ g of each cell lysate was fractionated by SDS-PAGE on a 10% polyacrylamide gel and transferred to a nitrocellulose membrane.
- the cell lysate was blocked for 1 hour at room temperature with 5% skim milk dissolved in tri-buffered saline (TBS) containing 0.02% Tween 20, and then reacted overnight at 4°C with a primary antibody (1:1000 dilution). Actin (1:5000 dilution) was used as a dose control.
- TBS tri-buffered saline
- the C2C12-PHF20/YY1-promoter-GFP cell line was treated with 50 ng/ml or 250 ng/ml of doxycycline, and after 24 hours, the degree of green luminescence was confirmed through a fluorescence microscope.
- the C2C12-PHF20/YY1-promoter-GFP cell line according to Example 1 was dispensed in a 96-well plate at 0.5 ⁇ 10 5 , cultured for 24 hours, and cells in a 70% confluent state were treated with 250 ng/ml of doxycycline for 24 hours, and then treated with each compound at a final concentration of 10 ⁇ M. After 24 hours of compound treatment, the degree of green luminescence was confirmed using GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA).
- C2C12-PHF20/YY1-promoter-GFP cells were dispensed at 4 ⁇ 10 5 in a 6-well culture plate and cultured for 24 hours. Then, cells in a 70% confluent state were treated with 250 ng/ml of doxycycline and then after 24 hours, treated with sulfamethazine, ciclopirox, or IOX1 for 24 hours at a final concentration of 10 ⁇ M, respectively. Thereafter, the degree of green luminescence was confirmed using GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA).
- 13 kinds of compounds having an effect of reducing green luminescence were derived as candidate substances, in which the compounds included sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide and sulfadiazine ( FIG. 4 ).
- the effect was confirmed using one of the candidate substances, sulfamethazine.
- the sulfamethazine was purchased from Sigma (catalog No.S8876) and used, and then dissolved in DMSO at 10 mM and subsequently used in cell experiments.
- the HTS system constructed in Example 1 was treated with 1 nM, 10 nM, 100 nM, 1 ⁇ M or 10 ⁇ M of sulfamethazine, respectively, to measure green fluorescence.
- the C2C12 muscle cell line was treated with each compound for each concentration for 24 hours, and the expression of proteins PHF20, YY1, and MyoD1 was confirmed by western blotting. In a group treated with each compound, it was confirmed that the protein expression of PHF20 and YY1 decreased, and the expression of MyoD increased ( FIG. 7 ).
- a sterile coverslip was placed in a 12-well plate, and 1 ⁇ 10 5 C2C12/Tet-On PHF20 cells per well were plated and grown at 37°C.
- the cells were treated with doxycycline for 24 hours.
- the cells were differentiated with DM (HS 2%) for 5 days, and at this time, each sulfonamide-based compound was treated for each concentration on day 4 of differentiation.
- DM HS 2%)
- each sulfonamide-based compound was treated for each concentration on day 4 of differentiation.
- the cells were washed twice with PBS at 37°C, reacted in 4% paraformaldehyde for 1 hour, and fixed on the coverslip. Then, the cells were washed twice with PBS.
- the cells were reacted with Triton x-100 in PBS for 30 minutes.
- the coverslip was blocked in 1% BSA for 1 hour at room temperature while shaking.
- An anti-Myosin (MF20) antibody was added to 1% BSA (1:200) and reacted overnight at 4 °C.
- the coverslip was washed three times each with PBS, and an Alexa Fluor 568 secondary antibody (1:1000) in 1% BSA was added, and then reacted at room temperature in the dark for 1 hour while mixing. Thereafter, the coverslip was washed three times with PBS and mounted on a slide using a mounting medium containing DAPI VECTASHIELD (St. Louis, USA), and images of the stained cells were photographed using Zeiss, which were illustrated in FIGS. 8A to 8C .
- FIG. 8A shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells after differentiation into myotubes from a group of treating a C2C12-PHF20 inducible cell line with doxycyclin to overexpress PHF20 and a control group treated with no doxycyclin, treating drugs (sulfamethazine, sulfatiazole, sulfadiazine, sulfadoxin, sulfadimethoxine, sulfacetamide sodium, sulfaphenazole, and sulfameter) on day 4 of differentiation, and then staining with heavy chain of myosin II (MF20) involved in muscle function and development on day 5 of differentiation.
- MF20 myosin II
- FIG. 8A shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfamethazine, sulfatiazole, sulfadiazine, sulfadoxin, sulfadimethoxine, sulfacetamide sodium, sulfaphenazole, and sulfameter
- FIG. 8B shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfamethazine, sulfapyridine, sulfanilamide, and sulfamethizole
- FIG. 8C shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfasalazine for each concentration.
- mice 32 48-week-old C57BL/6J male mice were obtained from KBSI and then bred under constant room temperature and 12 h night/day cycle for 2 weeks before the experiment, and the mice were fed with a standard rodent diet and freely taken with water.
- Treadmill adaptation was performed randomly for 3 days under conditions of grade 0, speed 15 (cm/sec), 10 minutes, electrical stimulation 02 AM (defined as exhaustion when electrical stimulation occurred consecutive 4 times or more), and according to the treadmill result, mice with similar ability were distributed into each group and divided into 4 groups of 8 mice per group.
- mice divided in 3-1 above were orally administered at 10:30 every day for 28 days with Group 1 of PBS (Hyclone Dulbecco's Phaosphate Buffer saline Cat. SH30028.02), Group 2 of sulfasalazine (Sigma, Cat. Nr S0883. CAS Number 599-79-1 ) 5 mpk, Group 3 of sulfasalazine 50 mpk, and Group 4 of sulfasalazine 500 mpk.
- PBS Hyclone Dulbecco's Phaosphate Buffer saline Cat. SH30028.02
- Group 2 of sulfasalazine Sigma, Cat. Nr S0883. CAS Number 599-79-1
- Group 3 of sulfasalazine 50 mpk Group 4 of sulfasalazine 500 mpk.
- mice were starved for 3 hours before measuring the treadmill, and then the treadmill was performed 4 times at 7-day intervals (0 days, 7 days, 14 days, and 28 days) according to Table 1 below.
- Table 1 Grade Time (min) Speed (cm/sec) 0 0 ⁇ 3 15 0 3 ⁇ 6 20 0 6 ⁇ 9 25 0 9 ⁇ 12 30 0 12 ⁇ 15 35 0 15 ⁇ 18 40 - Electrical stimulation 02 AM - Day 0 before administration, days 7, 14, 21, and 28 after administration - Defined as exhaustion when electric stimulation occurred consecutive 4 or more times
- the Rota Rod was measured on days 0, 7, 14, 21, and 28 with the animal group constructed in Group 2 under the conditions shown in Table 2 below, and the results were shown in FIG. 11 (( ⁇ ) p ⁇ 0.05 vs G1).
- the grip strength was measured on the 2 paws and 4 paws of the mouse on days 0, 9, 16, 23, and 28, and the results were shown in FIGS. 12A and 12B .
- FIGS. 11A and 11B it was confirmed that as the dose of sulfasalazine increased and the administration day increased, the recovery of balance ability of the mouse on the Rota Rod was improved.
- FIGS. 12A and 12B in both 2 paws and 4 paws measurement, it was confirmed that the grip strength was improved with the increase in sulfasalazine dose compared to a control group, Group 1, and the grip strength was improved as the administration day was increased.
- Group 1 of orally administering PBS to a high-fat diet mouse without injecting CTX Five groups of Group 1 of orally administering PBS to a high-fat diet mouse without injecting CTX, Group 2 of orally administering PBS to a high-fat diet mouse injected with CTX, Group 3 of orally administering sulfasalazine 5 mg/kg to the high-fat diet mouse injected with CTX, Group 4 of orally administering sulfasalazine 50 mg/kg to the high-fat diet mouse injected with CTX, and Group 5 of orally administering sulfasalazine 500 mg/kg to the high-fat diet mouse injected with CTX were divided and orally administered at 10 am every day for 14 days, and the body weights were measured and the results were shown in FIG. 13 .
- the left leg of an 8-week-old C57BL/6J male mouse was anesthetized with isoflurane, the entire leg was wound from the ankle with a 5 cm sports tape, and then wound and fixed with a Velcro with a width of 10 mm, and the Velcro condition was checked every day for 14 days ( FIG. 16 ).
- Velcro injury means that muscle damage is caused by such Velcro by winding the muscle with the Velcro so as not to move.
- mice For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed.
- mice with Velcro injury and mice without Velcro injury As illustrated in FIG. 18A , as a result of comparing distances run on the treadmill on the first day after unwinding the Velcro, it was confirmed that the mice wound with Velcro were injured with Velcro equally in each group (Group 2 to Group 5) compared to the mice unwound with Velcro (Group 1). As illustrated in FIG. 18B , the same result as the treadmill running distance was confirmed as compared with the treadmill running time.
- mice 7-week-old male and female C57BL/6J mice with the body weight of 20 ⁇ 3 g were obtained from DooYeol Biotech (Seoul, South Korea). The mice were bred under constant room temperature and 12 h night/day cycle for 1 week before the experiment, and the mice were fed with a standard rodent diet and were freely taken with water.
- cardiotoxin LATOXAN, Portes-lès-Valence, France
- PBS phosphatidylcholine
- the stock solution was diluted again in PBS at a final concentration of 0.03 mg/ml and used, and intramuscularly injected into both thigh muscles of the mouse using a 1 ml syringe (24G) to construct a sarcopenia mouse model.
- mice muscle tissue immunostaining the muscle tissues of a comparative group and a sulfamethazine-treated group were fixed in 4% paraformaldehyde, washed, and fixed in paraffin. Tissues were cut to a thickness of about 4 mm, fixed to a slide, and then stained using an antibody for each muscle type.
- mice For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed. As illustrated in FIG. 22 , no change in body weight was observed for each group during the entire experimental period, except for a decrease in body weight in all groups on day 10 without diet supply.
- mice For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed. As illustrated in FIG. 22 , no change in body weight was observed for each group during the entire experimental period, except for a decrease in body weight in all groups on day 10 without diet supply.
- mice 7-week-old male and female C57BL/6J mice with the body weight of 20 ⁇ 3 g were obtained from DooYeol Biotech (Seoul, South Korea). The mice were bred under constant room temperature and 12 h night/day cycle for 1 week before the experiment, and the mice were fed with a standard rodent diet and were freely taken with water. The experiment was conducted by dividing the mice into a control group and test groups (100 mg/kg, 500 mg/kg, and 1000 mg/kg), and sulfamethazine was diluted with 12.5% DMSO and 12.5% cremophor and orally administered at 10 ml/kg.
- the experiment was conducted for 2 weeks, and the general condition and the presence or absence of death were observed at least once a day.
- the body weights were measured before administration, and on 1 day, 3 days, 7 days, and 14 days after administration, and an autopsy was performed on day 14.
- a single oral dose toxicity test refers to a test that qualitatively and quantitatively examines toxicity in a short period of time when a test substance was administered to test animals in a single dose (including divided doses within 24 hours).
- FIG. 27A shows all mice for confirming toxicity according to single oral administration of female and male mice
- FIG. 27B shows photographs of male mice before organ harvesting
- FIG. 27C shows photographs of female mice before organ harvesting.
- FIG. 28A is a photograph confirming toxicity by dissecting the livers of male mice
- FIG. 28B is a photograph confirming toxicity by dissecting the livers of female mice
- FIG. 29A is a photograph confirming toxicity by dissecting the lungs of male mice
- FIG. 29B is a photograph confirming toxicity by dissecting the lungs of female mice.
- FIG. 30 is a photograph of confirming toxicity by dissecting the brains of male and female mice
- FIG. 31 is a photograph of confirming toxicity by dissecting the hearts of male and female mice
- FIG. 32 is a photograph of confirming toxicity by dissecting the stomachs of male and female mice
- FIG. 30 is a photograph of confirming toxicity by dissecting the brains of male and female mice
- FIG. 31 is a photograph of confirming toxicity by dissecting the hearts of male and female mice
- FIG. 32 is a photograph of confirming toxicity by dissecting the stomachs of male and female mice
- FIG. 33 is a photograph of confirming toxicity by dissecting the pancreases of male and female mice
- FIG. 34 is a photograph of confirming toxicity by dissecting the spleens of male and female mice.
- FIG. 35A is a photograph of confirming toxicity by dissecting the kidneys of male mice
- FIG. 35B is a photograph of confirming toxicity by dissecting the kidneys of female mice
- FIG. 36A is a photograph of confirming toxicity by dissecting the S. intestines of male mice
- FIG. 36B is a photograph of confirming toxicity by dissecting the S. intestines of female mice.
- FIG. 37A is a photograph of confirming toxicity by dissecting the L. intestines of male mice
- FIG. 37A is a photograph of confirming toxicity by dissecting the L. intestines of male mice
- FIG. 37B is a photograph of confirming toxicity by dissecting the L. intestines of female mice
- FIG. 38 is a photograph of confirming toxicity by dissecting the testis of male mice
- FIG. 39 is a photograph of confirming toxicity by dissecting the wombs of female mice.
- mice with single oral administration were dissected on day 14 after administration, and histochemical toxicity was evaluated.
- the pathology of the liver was observed through H&E staining on the dissected liver tissue.
- mice orally administered once as in Experimental Example 7-1 blood was collected from the heart of the mouse on day 14 after administration, and serum was collected to confirm the expression of a liver toxicity marker, a heart toxicity marker, and a kidney toxicity marker, respectively.
- blood samples were collected from the heart after surgery on day 14 after administration of sulfamethazine. The collected blood was left at room temperature for 1 hour to be coagulated, and the serum was isolated by operating a centrifuge (13,000 rpm) for 20 minutes. Blood analysis was performed by dividing liver toxicity, heart toxicity, and kidney toxicity, and the analysis was conducted by requesting DooYeol Biotech (Seoul, Korea).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Neurology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Physical Education & Sports Medicine (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Coloring Foods And Improving Nutritive Qualities (AREA)
- Fodder In General (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
Abstract
Description
- This application is based on and claims priority from
Korean Patent Application No. 10-2022-0043350, filed on April 07, 2022 Korean Patent Application No. 10-2022-0058865, filed on May 13, 2022 Korean Patent Application No. 10-2022-0186355, filed on December 27, 2022 - The present disclosure relates to a composition for improving, treating or preventing muscular disorders including sulfonamide-based compounds.
- Sarcopenia is a disease in which the muscle mass is reduced or the muscle strength decreases due to diseases, aging, and the like. The muscle mass gradually decreases after the age of 40's, and it is estimated that a decrease of 8% in the muscle mass occurs every 10 years until the age of 70's, and thereafter, a more rapid decrease in the muscle mass occurs, and it is known that the decrease in the muscle mass may occur up to 15% every 10 years.
- Through many follow-up studies, it has been found that the physiological changes that occur in the elderly are diverse, and in general, the muscle mass and the bone density decrease simultaneously with increasing age. Senile sarcopenia causes not only direct muscle strength reduction to increase the risk of death due to reduction and disability of various body functions, but also a decrease in metabolism and a decrease in immunity to increase the prevalence of metabolic diseases such as hypertension, diabetes, arthritis, obesity, and cancer. In particular, the senile sarcopenia is a disease that occurs in 40% of the elderly over 80 years of age and is expected to increase social and economic burdens in the aging era.
- PHD finger protein 20 (PHF 20) is a protein also known as glioma-expressed antigen 2 (GLEA2). Recently, it has been found that
PHF 20, as a transcription factor, acts on muscle damage and sarcopenia by regulating a sub-target protein YY1. It has been reported that the expression of PHF20 increases and then decreases during muscle cell differentiation using myoblasts (C2C12), and this phenomenon is regulated together with the expression of a muscle differentiation inhibitory protein YY1. - Meanwhile, there are three major treatment methods for sarcopenia. The first is exercise. It has been reported that the exercise increases the protein synthesis ability of skeletal muscle in the short term, and increases muscle strength or mobility in the elderly. However, the exercise is not suitable for long-term treatment. Second, testosterone or anabolic steroids can be used as drug treatment, but induces masculinization in women and causes side effects such as prostate symptoms in men. Other approved prescriptions include dehydroepiandrosterone (DHEA) and growth hormone, which have been reported in studies that can be used as treatment methods at sites containing selective androgen receptor modulators (SARMs). In addition, although diet therapy is known as a treatment method, according to nutritional assessments, malnutrition, and modern eating habits are inadequate to maintain adequate total body mass. However, the reality is that there is no fundamental therapeutic agent or enhancer for sarcopenia.
- In order to solve the problems, the present inventors of the present disclosure have screen materials that may restore or alleviate the inhibition of muscle differentiation under conditions that muscle differentiation in myoblasts is inhibited by PHF20 overexpression, by using a screening system constructed using PHF20/YY1. Accordingly, the present inventors confirmed that the selected compounds could be used to inhibit muscle reduction and promote muscle differentiation, and completed the present disclosure.
- According to an aspect of the present disclosure, there is provided a pharmaceutical composition for prevention or treatment that may help treat, relieve, alleviate or prevent diseases, including a sulfonamide-based compound represented by Chemical Formula 1 below or a salt thereof; or a food composition; a quasi-drug composition; a feed composition; or a composition for feed additives capable of preventing, improving, relieving or alleviating desired symptoms or assisting in the desired symptoms.
- In one embodiment, an objective of the present disclosure is to provide a pharmaceutical composition for preventing or treating muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 below or a pharmaceutically acceptable salt thereof.
- In one embodiment, another objective of the present disclosure is to provide a food composition for preventing or improving muscular disorders including a sulfonamide-based compound represented by Chemical Formula 1 below or a food acceptable salt thereof.
-
-
- In the present disclosure, the term "prevention" refers to all actions that inhibit the development of muscular disorders by administering the composition, or inhibit or delay the severity of muscular disorders.
- In the present disclosure, the term "treatment", "improvement", "relief", or "alleviation" refers to all actions that improve or beneficially change the symptoms of muscular disorders by administration of the composition.
- In the present disclosure, "capable of assisting" means to inhibit or delay symptoms of muscular disorders by administering the composition of the present disclosure, or to help or be able to help in improving the symptoms caused by muscular disorders. It may be used as an adjuvant to enhance an effect of medicines for treatment, and has meaning as an adjuvant as a health functional food or functional food.
-
- The terms used in the exemplary embodiments are used for the purpose of description only, and should not be construed to be limited. The singular expression includes the plural expression unless the context clearly dictates otherwise. In the present application, it should be understood that term "comprising" or "having" indicates that a feature, a number, a step, an operation, a component, a part or the combination thereof described in the specification is present, but does not exclude a possibility of presence or addition of one or more other features, numbers, steps, operations, components, parts or combinations thereof, in advance.
- Unless otherwise contrarily defined, all terms used herein including technological or scientific terms have the same meanings as those generally understood by a person with ordinary skill in the art to which exemplary embodiments pertain. Terms which are defined in a generally used dictionary should be interpreted to have the same meaning as the meaning in the context of the related art, and are not interpreted as ideal or excessively formal meanings unless otherwise defined in the present application.
- In addition, in the description with reference to the accompanying drawings, like components designate like reference numerals regardless of reference numerals and a duplicated description thereof will be omitted. In describing the example exemplary embodiments, a detailed description of related known technologies will be omitted if it is determined that they unnecessarily make the gist of the example exemplary embodiments unclear.
- According to the present disclosure, the composition for preventing, improving or treating muscular disorders including the sulfonamide-based compounds or salts thereof can prevent inhibition of differentiation of myoblasts by regulating the expression of PHF20 and YY1. Accordingly, since the composition may prevent or alleviate muscle loss, or promote muscle regeneration to improve muscle functions and muscle mass, the composition can be effectively used for therapeutic agents, foods, or feeds for prevention, improvement or treatment of muscle disorders, and improving muscle functions or muscle mass.
- The foregoing summary is illustrative only and is not intended to be in any way limiting. In addition to the illustrative aspects, embodiments, and features described above, further aspects, embodiments, and features will become apparent by reference to the drawings and the following detailed description.
-
-
FIG. 1 is a diagram showing a process of constructing a high throughput screening (HTS) system in a cell line that may easily monitor the differentiation pattern of muscle cells according to an exemplary embodiment of the present disclosure. -
FIGS. 2A and2B are diagrams illustrating results of confirming PHF20 expression (FIG. 2A ) and GFP expression (FIG. 2B ) according to a concentration of a selection marker in order to prepare a stable cell line for constructing a high throughput screening (HTS) system according to an exemplary embodiment of the present disclosure. -
FIG. 3 is a fluorescent picture and a graph showing a result of GFP expression according to a marker concentration and time in order to prepare a stable cell line for constructing a high throughput screening (HTS) system according to an exemplary embodiment of the present disclosure. -
FIG. 4 is a diagram illustrating a result of screening candidate substances using the HTS system constructed according to an exemplary embodiment of the present disclosure. -
FIG. 5 is a graph showing an effect of suppressing an action of inhibiting the formation of myoblasts upon treatment with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 6 is a graph showing IC50 values based on the results of suppressing the inhibition of the formation of myoblasts by concentration of sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 7 is a diagram illustrating a Western blot analysis result for each compound in a C2C12 muscle cell line, according to an exemplary embodiment of the present disclosure. -
FIGS. 8A to 8C are cellular immunofluorescence staining images for confirming the differentiation ability of muscle cells after differentiation into myotubes from a group of treating a C2C12-PHF20 inducible cell line with doxycyclin to overexpress PHF20 and a control group treated with no doxycyclin, treating a drug (sulfonamide-based compound) onday 4 of differentiation, and then staining with heavy chain of myosin II (MF20) involved in muscle function and development onday 5 of differentiation, according to an exemplary embodiment of the present disclosure. -
FIG. 9 is a graph of confirming a change in body weight of mice for each administration group in order to confirm a treadmill exercise capacity recovery effect depending on a dose of sulfasalazine in an aging mouse model according to an exemplary embodiment of the present disclosure (a horizontal axis of the graph means a day of administration, and a vertical axis means a weight of the mouse). -
FIG. 10 is a graph showing changes in treadmill exercise capacity recovery effect depending on a dose of sulfasalazine in an aging mouse model according to an exemplary embodiment of the present disclosure (a horizontal axis of the graph means a day of administration, and a vertical axis means a latency to fall). -
FIG. 11 is a graph showing changes in balance ability recovery effect of a Rota Rod depending on a dose of sulfasalazine in an aging mouse model according to an exemplary embodiment of the present disclosure. -
FIGS. 12A and 12B are diagrams illustrating confirming a grip strength recovery effect depending on a dose of sulfasalazine in an aging mouse model according to an exemplary embodiment of the present disclosure, and show changes in grip strength recovery (12A) of hind legs (2 paws) and changes in grip strength recovery (12B) of all legs (4 paws) depending on a dose. -
FIG. 13 is a graph of confirming changes in body weight of mice for each administration group in order to confirm a treadmill exercise capacity recovery effect depending on a dose of sulfasalazine in a high-fat diet CTX sarcopenia mouse model according to an exemplary embodiment of the present disclosure. -
FIG. 14 is a graph showing changes in balance ability recovery effect of a Rota Rod depending on a dose of sulfasalazine in a high-fat diet CTX sarcopenia mouse model according to an exemplary embodiment of the present disclosure. -
FIGS. 15A and 15B are diagrams illustrating confirming a grip strength recovery effect depending on a dose of sulfasalazine in a high-fat diet CTX sarcopenia mouse model according to an exemplary embodiment of the present disclosure and show changes in grip strength recovery (15A) of hind legs (2 paws) and changes in grip strength recovery (15B) of all legs (4 paws) depending on a dose. -
FIG. 16 is an image of photographing a Velcro injured sarcopenia mouse model according to an exemplary embodiment of the present disclosure, in which a left image is an image taken after winding a sports tape around the ankle and calf of the mouse, and middle and right images are images taken after winding and fixing a sports tape around the entire leg from the ankle of the mouse and then winding and fixing the entire leg with Velcro. -
FIG. 17 is a graph of confirming changes in body weight of mice for each administration group in order to confirm a treadmill exercise capacity recovery effect depending on a dose of sulfasalazine in a Velcro injured sarcopenia mouse model according to an exemplary embodiment of the present disclosure. -
FIGS. 18A and18B are diagrams showing a distance (FIG. 18A ) and time (FIG. 18B ) of running a treadmill before drug administration in order to determine whether to construct a Velcro injured sarcopenia mouse model according to an exemplary embodiment of the present disclosure (inFIG. 18A , a horizontal axis means a relative ratio (%) to a distance in a control group, and a vertical axis means a drug treatment group, and inFIG. 18B , a horizontal axis means a relative ratio (%) to time in a control group, and a vertical axis means a drug treatment group). -
FIGS. 19A and19B are graphs of confirming distance (FIG. 19A ) and time (FIG. 19B ) run on a treadmill for each group depending on a dose and an administration date of sulfasalazine in order to confirm treadmill exercise capacity of a Velcro injured sarcopenia mouse model according to an exemplary embodiment of the present disclosure (inFIG. 19A , a horizontal axis means an administration day, and a vertical axis means a distance, and inFIG. 19B , a horizontal axis means an administration day, and a vertical axis means time). -
FIG. 20 is a photograph of muscle damage recovery observed with an optical microscope after H&E staining after oral administration of sulfamethazine for 10 days after CTX injection into the thigh of a mouse according to an exemplary embodiment of the present disclosure. -
FIG. 21 is a photograph observed under a fluorescence microscope to identify a muscle recovery effect for each muscle type (myosin type I, myosin type IIa, and myosin type IIb) according to oral administration of sulfamethazine for 10 days in a CTX-induced sarcopenia mouse model according to an exemplary embodiment of the present disclosure. -
FIG. 22 is a graph of confirming changes in body weight of mice for each administration group in order to confirm a balance ability recovery effect of Rota Rod according to a dose of sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 23 is a diagram illustrating a method and conditions for confirming Rota Rod balance ability according to an exemplary embodiment of the present disclosure. -
FIG. 24 is a graph showing changes in balance ability recovery effect of a Rota Rod depending on a dose of sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 25 is a graph of confirming changes in body weight of mice for each administration group in order to confirm a grip strength recovery effect according to a dose of sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 26A and26B are diagrams illustrating confirming a grip strength recovery effect depending on a dose of sulfamethazine according to an exemplary embodiment of the present disclosure, and show changes in grip strength recovery (26A) of hind legs (2 paws) and changes in grip strength recovery (26B) of all legs (4 paws) depending on a dose. -
FIGS. 27A to 27C are diagrams illustrating results of confirming toxicity according to a single oral administration of sulfamethazine according to an exemplary embodiment of the present disclosure:- Control - PBS administration;
- Group 1 -
Sulfamethazine 100 mg/kg; - Group 2 -
Sulfamethazine 500 mg/kg; - Group 3 -
Sulfamethazine 1000 mg/kg)
-
FIGS. 28A and28B are photographs of confirming toxicity to the liver in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 29A and29B are photographs of confirming toxicity to the lung in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 30 is a photograph of confirming toxicity to the brain in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 31 is a photograph of confirming toxicity to the heart in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 32 is a photograph of confirming toxicity to the stomach in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 33 is a photograph of confirming toxicity to the pancreas in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 34 is a photograph of confirming toxicity to the spleen in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 35A and 35B are photographs of confirming toxicity to the kidney in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 36A and 36B are photographs of confirming toxicity to the small intestine in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 37A and 37B are photographs of confirming toxicity to the large intestine in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 38 is a photograph of confirming toxicity to the testis in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 39 is a photograph of confirming toxicity to the womb in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIG. 40 is a graph showing changes in body weight for each administration group in mice orally administered with sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 41A and41B are graphs of confirming a weight of each of the brain, lung, heart, liver, pancreas, stomach, spleen, kidney, small intestine, large intestine, testis and womb of each administration group in mice onday 14 after orally administering sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 42A and42B are photographs showing H&E staining to confirm toxicity in the liver of mice onday 14 after orally single-administering sulfamethazine according to an exemplary embodiment of the present disclosure. -
FIGS. 43A and43B are graphs showing confirming the expression of blood liver toxicity, heart toxicity, and kidney toxicity markers in mice orally single-administered with sulfamethazine according to an exemplary embodiment of the present disclosure. - Hereinafter, the present disclosure will be described in more detail.
- The present disclosure provides a use for preventing, improving or treating muscular disorders including a sulfonamide-based compound represented by
Chemical Formula 1 or a salt thereof; and a composition for preventing, improving or treating muscular disorders including a sulfonamide-based compound represented byChemical Formula 1 or a salt thereof. -
- In one embodiment, the hetero element of the hetero aryl may be one or more selected from S, N, and O, one or more hetero elements may be included, and one or more hetero elements different from each other may be included.
- In one embodiment, the aryl or heteroaryl may be selected from the group consisting of pyrrole, pyrazole, triazole, oxazole, furan, isoxazole, isothiazole, imidazole, diazole, thiadiazole, phenyl, pyridine, pyrimidine, triazine, oxazine, and thiazine.
- In one embodiment, the aryl or heteroaryl may be thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine.
- In one embodiment, the substituent of R1 may be at least one substituent selected from C1-3 alkyl, C1-3 alkoxy, and phenyl. In a preferred embodiment, the substituent of R1 may be at least one selected from the group consisting of methyl, ethyl, methoxy, ethoxy and phenyl. One substituent of R1 may be included, and one or more substituents identical to or different from each other may be included.
- In one embodiment, the compound of
Chemical Formula 1 may be sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide or sulfadiazine. - In one embodiment, the salt of the compound of the present disclosure includes all kinds of salts commonly accepted in the art of the present disclosure, and may be, for example, a sodium salt.
-
-
-
-
-
-
-
-
-
-
-
-
-
- In the present disclosure, the muscular disorder means a disease or symptom that causes a reduced muscle function; a decrease in muscle mass; muscle wasting; or muscle degeneration, or worsens these symptoms.
- In this specification, the term 'muscle' collectively refers to tendons and muscles. Further, the `muscle function' refers to the ability to exert power by contraction of muscles, and includes muscular strength which is the ability of the muscles to exhibit maximum contraction to overcome resistance, muscular endurance which is the ability to exhibit how long or how many times muscles may repeat contraction and relaxation at a given weight, and quickness which is the ability to exert strong power in a short time.
- In this specification, the term `improvement of muscle function' refers to improving the muscle function better by increasing the muscle mass, promoting regeneration of damaged muscles, or improving regeneration.
- In this specification, the `promoting the muscle regeneration' means shortening the regeneration or recovery time of damaged muscles, increasing muscle mass by activating the muscles, or lowering the degree of muscle mass loss.
- The muscle wasting and degeneration are caused by genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle, but is not limited to a type of muscle.
- In one embodiment, the muscular disorder may be one selected from the group consisting of sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia.
- The present disclosure provides a pharmaceutical composition for preventing or treating muscular disorders including a sulfonamide-based compound represented by
Chemical Formula 1 or a salt thereof; a food composition for preventing or treating muscular disorders; and a feed composition for preventing or treating muscular disorders: -
- In one embodiment, the hetero element of the hetero aryl may be one or more selected from S, N, and O, one or more hetero elements may be included, and one or more hetero elements different from each other may be included.
- In one embodiment, the aryl or heteroaryl may be selected from the group consisting of pyrrole, pyrazole, triazole, oxazole, furan, isoxazole, isothiazole, imidazole, diazole, thiadiazole, phenyl, pyridine, pyrimidine, triazine, oxazine, and thiazine.
- In one embodiment, the aryl or heteroaryl may be thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine.
- In one embodiment, the substituent of R1 may be at least one substituent selected from C1-3 alkyl, C1-3 alkoxy, and phenyl. In a preferred embodiment, the substituent of R1 may be at least one selected from the group consisting of methyl, ethyl, methoxy, ethoxy and phenyl. One substituent of R1 may be included, and one or more substituents identical to or different from each other may be included.
- In one embodiment, the compound of
Chemical Formula 1 may be sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide or sulfadiazine. - In one embodiment, the salt of the compound of the present disclosure includes all kinds of salts commonly accepted in the art of the present disclosure, and may be, for example, a sodium salt.
- In one embodiment, the compound of
Chemical Formula 1 of the present disclosure acts on PHD20/YY1 to suppress the inhibition of the formation of myoblasts, thereby inhibiting a decrease in muscles in muscle tissues or promoting muscle formation. - As an embodiment, the sulfamethazine of the present disclosure acts on PHD20/YY1 to suppress the inhibition of the formation of myoblasts, thereby inhibiting muscle damage and promoting regeneration, and as a result, it was confirmed in vivo that the balance ability and grip strength recovery of Rota Rod were superior to those of muscle-damaged mice.
- When the composition for preventing or treating the muscular disorders of the present disclosure is the pharmaceutical composition, the composition may be used for preventing or treating muscular disorders caused by muscle wasting or degeneration. The muscle wasting and degeneration occur due to genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle. Examples of related disorders may include sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia, but are not limited thereto. The pharmaceutical composition of the present disclosure may include a pharmaceutically acceptable carrier.
- The pharmaceutically acceptable carrier may further include, for example, a carrier for oral administration or a carrier for parenteral administration. The carrier for oral administration may include lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. In addition, the carrier for parenteral administration may include water, suitable oil, saline, aqueous glucose, glycol, and the like. In addition, a stabilizer and a preservative may be further included. A suitable stabilizer includes antioxidants such as sodium hydrogen sulfite, sodium sulfite or ascorbic acid. A suitable preservative includes benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol.
- The pharmaceutical composition of the present disclosure may be administered to mammals including humans even by any method. For example, the pharmaceutical composition may be administered orally or parenterally, and the parenteral administration method is not limited thereto, but may include intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intranasal, intestinal, topical, sublingual or rectal administration.
- The pharmaceutical composition of the present disclosure may be formulated as formulations for oral administration or parenteral administration according to the route of administration as described above. The formulation may be prepared by using one or more buffers (e.g., saline or PBS), antioxidants, bacteriostatic agents, chelating agents (e.g., EDTA or glutathione), fillers, extenders, binders, adjuvants (e.g., aluminum hydroxide), suspending agents, thickening agents, wetting agents, disintegrants or surfactants, and diluents or excipients.
- Solid preparations for oral administration include tablets, pills, powders, granules, liquids, gels, syrups, slurries, suspensions or capsules. These solid preparations may be prepared by mixing at least one excipient, for example, starch (including corn starch, wheat starch, rice starch, potato starch, etc.), calcium carbonate, sucrose, lactose, dextrose, sorbitol, mannitol, xylitol, erythritol maltitol, cellulose, methyl cellulose, sodium carboxymethyl cellulose and hydroxypropyl methyl cellulose or gelatin with the pharmaceutical composition of the present disclosure. For example, tablets or sugar-coated tablets may be obtained by mixing an active ingredient with a solid excipient, pulverizing the mixture, adding a suitable adjuvant, and then processing the mixture into a granule mixture.
- Lubricants such as magnesium stearate and talc is also used in addition to simple excipients. Liquid preparations for oral administration may correspond to suspensions, oral liquids, emulsions, syrups, or the like, and may include various excipients, for example, a wetting agent, a sweetener, an aromatic agent, and a preserving agent, in addition to water or liquid paraffin which is a commonly used simple diluent. In addition, in some cases, cross-linked polyvinylpyrrolidone, agar, alginic acid or sodium alginate may be added as a disintegrant, and an anti-coagulant, a lubricant, a wetting agent, a fragrance, an emulsifier, a preservative, and the like may be additionally included.
- As one embodiment, when administered parenterally, the pharmaceutical composition of the present disclosure may be formulated according to a method known in the art in the form of injections, transdermal agents, and nasal inhalers together with suitable parenteral carriers. The injections need to be sterilized and protected from contamination of microorganisms such as bacteria and fungi. Examples of suitable carriers for injections are not limited thereto, but may be solvents or dispersion media containing water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols), mixtures thereof and/or vegetable oils. More preferably, as suitable carriers, a Hanks' solution, a Ringer's solution, a phosphate buffered saline (PBS) containing triethanol amine or sterile water for injection, and an isotonic solution such as 10% ethanol, 40% propylene glycol and 5% dextrose may be used. In order to protect the injection from microbial contamination, various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid, and thimerosal may be further included. In addition, most of the injections may further include an isotonic agent, such as sugar or sodium chloride.
- As an example, the transdermal agents are included in the form of ointments, creams, lotions, gels, external liquids, pastas, liniments, and aerosols. Hereinabove, the `transdermal administration' means that an effective dose of the active ingredient contained in the pharmaceutical composition is delivered into the skin by topically applying the pharmaceutical composition to the skin.
- As an example, in the case of the inhalers, the compound used according to the present disclosure may be conveniently delivered in the form of an aerosol spray from a pressurized pack or a nebulizer by using a suitable propellant, for example, dichlorofluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, and carbon dioxide, or other suitable gases. In the case of a pressurized aerosol, a dosage unit may be determined by providing a valve that delivers a metered amount. For example, gelatin capsules and cartridges used in an inhaler or insufflator may be formulated to contain a powder mixture of a compound and a suitable powder base such as lactose or starch.
- The pharmaceutical composition for preventing or treating the muscular disorders of the present disclosure may provide preferred preventive or therapeutic effects on muscular disorders when including an effective dose of the compound of
Chemical Formula 1. - In one embodiment, there is provided a method for treating muscular disorders, including administering the compound represented by
Chemical Formula 1 or a pharmaceutically acceptable salt thereof to a subject in a therapeutically effective dose. - Preferably, the treatment method may further include identifying a subject in need of prevention or treatment of the muscular disorders before the administering step.
- In the present specification, the `therapeutically effective dose' or `effective dose' refers to an amount that exhibits a greater response than that of a negative control group, and preferably refers to an amount sufficient to improve muscle function. The pharmaceutical composition of the present disclosure may contain 0.01 to 99.99% of the compound of
Chemical Formula 1 or the pharmaceutically acceptable salt thereof, and the remaining amount may account for the pharmaceutically acceptable carrier. The effective dose of the compound ofChemical Formula 1 or the pharmaceutically acceptable salt thereof included in the pharmaceutical composition of the present disclosure may vary depending on a form in which the composition is commercialized. - The total effective dose of the pharmaceutical composition of the present disclosure may be administered to a subject in a single dose, or may be administered to the subject in a multiple dose for a long period of time according to a fractionated treatment protocol. In the pharmaceutical composition of the present disclosure, the content of the active ingredient may vary depending on the severity of disease. In the dosage, an effective dose to the subject may be determined by considering various factors such as the age, weight, health condition, sex, severity of disease, diet and excretion rate of the subject as well as the route of administration of the pharmaceutical composition and the number of treatments. Considering these points, those skilled in the art may determine an appropriate effective dose of the compound of
Chemical Formula 1 or the pharmaceutically acceptable salt thereof according to a specific use for preventing or treating muscular disorders. The pharmaceutical composition according to the present disclosure is not particularly limited to the formulation, the administration route, and the administration method thereof, as long as the effects of the present disclosure are shown. - The "subject" may refer to mammals such as human or non-human primates, mice, dogs, cats, horses, and cows, but is not limited thereto.
- The pharmaceutical composition for preventing or treating the muscular disorders of the present disclosure may also be provided in a formulation for external use including the compound of
Chemical Formula 1 or the pharmaceutically acceptable salt thereof as an active ingredient. - When the pharmaceutical composition of the present disclosure is used as an external skin preparation, the pharmaceutical composition may further contain adjuvants commonly used in the field of dermatology, such as any other ingredients commonly used in external skin preparations, such as fatty substances, organic solvents, solubilizers, concentrate and gelling agents, emollients, antioxidants, suspending agents, stabilizers, foaming agents, fragrances, surfactants, water, ionic emulsifiers, nonionic emulsifiers, fillers, sequestering agents, chelating agents, preservatives, vitamins, blockers, wetting agents, essential oils, dyes, pigments, hydrophilic activators, lipophilic activators or lipid vesicles. In addition, the ingredients may be introduced in an amount generally used in the field of dermatology.
- When the pharmaceutical composition of the present disclosure is provided as an external skin preparation, the external skin preparation is not limited thereto, but may be a formulation such as ointments, patches, gels, creams, or sprays.
- In another aspect, a composition for preventing or improving muscular disorders of the present disclosure may be a food composition for preventing or improving muscular disorders including a compound of
Chemical Formula 1 or a food acceptable salt thereof. - When the composition for preventing or treating the muscular disorders of the present disclosure is the food composition, the composition may be used for preventing or treating muscular disorders caused by muscle wasting or degeneration. The muscle wasting and degeneration occur due to genetic factors, acquired factors, aging, etc., and the muscle wasting is characterized by a gradual loss of muscle mass, weakness and degeneration of muscles, especially skeletal or voluntary muscle and cardiac muscle. Examples of related disorders may include sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia, but are not limited thereto.
- As one embodiment, the food composition means including all forms such as functional foods, nutritional supplements, health foods, food additives, or feeds, and is taken for animals including humans or livestock.
- The type of food composition may be prepared in various forms according to a general method known in the art. General foods are not limited thereto, but may be prepared by adding the compound of
Chemical Formula 1 of the present disclosure or the food acceptable salt thereof to beverages (including alcoholic beverages), fruits and processed foods thereof (e.g., canned fruit, bottled food, jam, and marmalade), fish, meat and processed foods thereof (e.g. ham, sausage, and corned beef), bread and noodles (e.g. udon, buckwheat noodles, ramen, spaghetti, and macaroni), fruit juice, various drinks, cookies, sweets, dairy products (e.g. butter, and cheese), edible vegetable oil, margarine, vegetable proteins, retort food, frozen food, various seasonings (e.g., soybean paste, soy sauce, and sauce), etc. In addition, the nutritional supplements are not limited thereto, but may be prepared by adding the compound ofChemical Formula 1 of the present disclosure or the food acceptable salt thereof to capsules, tablets, pills, etc. In addition, the health functional food is not limited thereto, but for example, may be taken by liquefying, granulating, encapsulating, and powdering so as to be drunken (health beverages) by preparing the compound ofChemical Formula 1 of the present disclosure or the food acceptable salt thereof in the form of tea, juice, and drinks. - In addition, in order to use the compound of
Chemical Formula 1 of the present disclosure or the food chemically acceptable salt thereof in the form of food additives, the health functional food may be prepared and used in the form of powders or concentrates. In addition, the health functional food may be prepared in the form of a composition by mixing the compound ofChemical Formula 1 of the present disclosure or the food acceptable salt thereof with known active ingredients known to have an effect of preventing or improving muscular disorders. - When the composition for preventing or improving muscular disorders of the present disclosure is used as a health drink composition, the health drink composition may contain various flavoring agents or natural carbohydrates as an additional ingredient, like a general drink. The natural carbohydrates may be monosaccharides such as glucose and fructose; disaccharides such as maltose and sucrose; polysaccharides such as dextrin and cyclodextrin; and sugar alcohols such as xylitol, sorbitol, and erythritol. As the sweetening agent, natural sweetening agents such as thaumatin and stevia extracts; synthetic sweetening agents such as saccharin and aspartame may be used. A ratio of the natural carbohydrates may be generally about 0.01 to 0.04 g, preferably about 0.02 to 0.03 g per 100 mL of the composition of the present disclosure.
- The compound represented by
Chemical Formula 1 of the present disclosure or the food acceptable salt thereof may be included as an active ingredient in the food composition for preventing or improving the muscular disorders, but the amount is not particularly limited to an amount effective to achieve an action for preventing or improving muscular disorders, but is preferably 0.01 to 100 wt% based on the total weight of the total composition. The food composition of the present disclosure may be prepared by mixing the compound represented byChemical Formula 1 or the food acceptable salt thereof with other active ingredients known to be effective in the composition for preventing or improving the muscular disorders. - In addition, the food composition may contain various nutrients, vitamins, electrolytes, flavoring agents, coloring agents, pectic acid and salts thereof, alginic acid and salts thereof, organic acid, protective colloidal thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohol, carbonic acid agents, or the like. In addition, the health food of the present disclosure may contain pulp for preparing natural fruit juice, fruit juice beverage, or vegetable beverage. These ingredients may be used independently or in combination. Although the ratio of these additives is not very important, generally, the ratio thereof is selected in a range of 0.01 to 0.1 part by weight per 100 parts by weight of the composition of the present disclosure.
- In yet another aspect, the composition for preventing or improving muscular disorders of the present disclosure may be a feed composition or a feed additive composition for preventing or improving muscular disorders including the compound of
Chemical Formula 1. - In the present disclosure, the term 'feed' may mean any natural or artificial diet, one-meal diet, or ingredients of one-meal diet to be eaten, ingested, and digested by animals or suitable therefor. The feed including the composition for preventing or improving the muscular disorders according to the present disclosure as an active ingredient may be prepared with various types of feed known in the art, and preferably, may include concentrated feeds, roughage and/or special feeds, but is not limited thereto.
- In the present disclosure, the term `feed additives' includes substances added to the feeds for the purpose of various effects, such as nutrient supplementation and weight loss prevention, improvement of digestibility of fiber in feed, oil quality improvement, reproduction disorder prevention and conception rate improvement, and summer high temperature stress prevention. The feed additives composition of the present disclosure correspond to supplementary feeds under the Feed Management Act, and may further include mineral preparations such as sodium bicarbonate, bentonite, magnesium oxide, and complex minerals, mineral preparations which are trace minerals such as zinc, copper, cobalt, and selenium, vitamins such as carotene, vitamins A, D, E, nicotinic acid, and vitamin B complex, protected amino acid supplements such as methionine and lysine, protected fatty acid preparations such as fatty acid calcium salts, active bacteria such as probiotics (lactic acid bacteria), yeast cultures, and mold fermented products, yeast agents, and the like.
- The concentrated feeds include seed fruits including grains such as wheat, oats and corn, bran including rice bran, wheat bran, and barley bran as by-products obtained by refining grains, residues such as seed cakes which are by-products obtained from oil extraction of beans, oil, sesame, linseed, and coco palms, and residual starches as a main ingredient of starch residues which are the remainder after removing starch from sweet potatoes and potatoes, fish soluble which is a concentrate of fresh liquids obtained from fish meal, fish residues, and fish, animal feeds such as meat meal, blood meal, feather meal, skim milk powder, dried whey, which is obtained by drying whey as the balance when preparing cheese from milk and casein from skim milk, yeast, chlorella, and seaweed, but are not limited thereto.
- The roughage includes raw grass feed such as wild grass, grass, and green cutting, root vegetables such as turnips for feed, beets for feed, and Lutherbearer as a type of turnip, silage which is stored feed made by filling a silo with green grass, green crops, and grains, and fermenting the silo with lactic acid, dried hay made by cutting and drying wild grass and grass, straw of breeding crops, and leaves of leguminous plants, but is not limited thereto. The special feed includes mineral feeds such as oyster shells and rock salt, urea feeds such as urea or its derivative, diureideisobutane, feed additives which are substances supplemented with ingredients that tend to be insufficient when only natural feed raw materials are mixed, or added in a small amount to the formulated feed to improve the storage of feeds, and dietary supplements, but is not limited thereto.
- The feed additives for preventing or improving the muscular disorders according to the present disclosure may be prepared by adding the compound of
Chemical Formula 1 of the present disclosure or the salt thereof in an appropriate effective concentration range according to various feed preparation methods known in the art. - In order to avoid redundant description, the definitions and descriptions of each component of the present disclosure described in
Chemical Formula 1 and the pharmaceutical composition section are applied to the food composition and the feed composition as they are, unless otherwise specified. - Hereinafter, exemplary embodiments will be described in detail with reference to the accompanying drawings. However, since various modifications may be made to the exemplary embodiments, the scope of the present disclosure is not limited or restricted by these exemplary embodiments. It should be understood that all modifications, equivalents and substitutes for the exemplary embodiments are included in the scope of the present disclosure.
- In order to find a sarcopenia control substance, a high throughput screening (HTS) system was constructed in a cell line capable of easily monitoring a differentiation pattern of muscle cells.
- Specifically, a mouse normal myoblast C2C12 cell line was obtained from ATCC (Manassas, VA, USA). C2C12 cells were subcultured every 2 to 3 days in a 100 mm dish containing a DMEM supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 unit/ml penicillin and 100 µg/ml streptomycin (Life Technologies, Grand Island, NY, USA). The cells were cultured and grown under 5% CO2 and 37°C conditions in a humidified state. While used in experiments, the cells were collected at the end of treatment for additional analysis.
- C2C12 myoblasts, which were stable cells in which PHF20 was overexpressed, were transformed with a plasmid coexisting with a YY1 promoter and GFP, which inhibited muscle differentiation, to prepare a stable cell line. The degree of GFP signal suppression was quantified and measured, and the degree of muscle differentiation was used as a quantitative measurement index (
FIG. 1 ). - In order to construct an HTS system capable of being used as a measurement index of a quantitative measurement system of the degree of muscle differentiation, a stable cell line was selected by treatment with hygromycin, a selection marker for GFP expression, by concentration.
- Specifically, C2C12 cell lines transformed with PHF20 and YY1-promoter-GFP plasmid were treated with 150 µg/ml (1 set), 50 µg/ml (2 set) or 250 µg/ml (3 set) of hygromycin together with 1 mg/ml of neomycin and 2 µg/ml of puromycin, respectively, to select only perfectly transformed cell lines. The medium was replaced once every two days, and neomycin, puromycin, and hygromycin were treated at each concentration, and proceeded for about a month.
- In addition, C2C12-PHF20/YY1-promoter-GFP cell lines of three types (150 µg/ml (1 set), 50 µg/ml (2 set) or 250 µg/ml (3 set)) selected separately were dispensed in a 96-well white plate in 0.5 × 105, cultured for 24 hours, and cells in a 70% confluent state were treated with doxycycline at 0, 50 ng/ml, 250 ng/ml, 500 ng/ml, or 1000 ng/ml, and then cultured for 24 hours. In order to confirm the fluorescence of GFP according to a concentration of doxycycline, the cells were collected and the fluorescence was confirmed using GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA). Extension values were set to 435 to 488 and then measured to quantify the luminescence degree of GFP.
- In addition, for Western blot analysis, C2C12 cells were left on ice, washed twice with cold PBS, an then dissolved at 4°C for 30 minutes in a cell lysis buffer (50 mmol/L Tris-HCl, pH 7.5, 1% (v/v) Nonidet P-40, 250 mmol/L NaCl, 0.1 mmol/L phenyl methyl sulfonyl fluoride, 0.1 mmol/L sodium vanadate, 20 mmol/L β-glycerol phosphate, 2 mmol/L DTT, 1 mmol/L Leupeptin and 10 mmol/L PNPP). Thereafter, the cell lysate was centrifuged at 16,000× g at 4°C for 20 minutes. The supernatant was collected and used for SDS-PAGE, and the protein content was evaluated by bovine serum albumin protein assay. Proteins were mixed with a sample buffer containing β-mercamptoethanol and heated at 100°C for 2 minutes. 40 µg of each cell lysate was fractionated by SDS-PAGE on a 10% polyacrylamide gel and transferred to a nitrocellulose membrane. The cell lysate was blocked for 1 hour at room temperature with 5% skim milk dissolved in tri-buffered saline (TBS) containing 0.02
% Tween 20, and then reacted overnight at 4°C with a primary antibody (1:1000 dilution). Actin (1:5000 dilution) was used as a dose control. After the cell lysate was reacted with the primary antibody, before the cell lysate was dissolved in TBS/Tween-20 containing 5% skim milk and reacted for 1 hour at room temperature with goat anti-mouse or anti-rabbit horse radish peroxidase (HRP)-conjugated antibody diluted at 1:2000, blots were washed 4 times in TBS/Tween-20. After washing in TBS/Tween-20, the blots were used for antigen detection using an enhanced chemiluminescence system. Proteins were visualized with an ECL-chemiluminescence kit (GE Healthcare, Life Sciences). - As illustrated in
FIGS. 2A and2B , as a result of confirming using a fluorescence microscope and Western blot analysis in the cell line in Example 1-1, it was confirmed that the luminescence intensity was increased in a doxycycline-dose dependent manner. As a result of comparative analysis by collecting the degree of light luminescence under each condition, the condition of treating 250 µg/ml of hygromycin was the most efficient, and the experiment was performed by fixing the condition of treating 250 ng/ml of doxycycline. - The C2C12-PHF20/YY1-promoter-GFP cell line was treated with 50 ng/ml or 250 ng/ml of doxycycline, and after 24 hours, the degree of green luminescence was confirmed through a fluorescence microscope.
- In addition, the degree of green luminescence was confirmed using a GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA) after treating the C2C12-PHF20/YY1-promoter-GFP cell line with 250 ng/ml of doxycycline for 12, 24, 36, and 48 hours, respectively.
- As a result, as illustrated in
FIG. 3 , it was confirmed that the degree of luminescence increased as the reaction time increased under a fixed doxycycline concentration condition (250 ng/ml). - In order to screen substances using the HTS system constructed in Example 1, an FDA-approved drug library (APExBIO, DiscoveryProbe) was purchased and the degree of GFP inhibition for a total of 1670 compounds was confirmed.
- Specifically, the C2C12-PHF20/YY1-promoter-GFP cell line according to Example 1 was dispensed in a 96-well plate at 0.5 × 105, cultured for 24 hours, and cells in a 70% confluent state were treated with 250 ng/ml of doxycycline for 24 hours, and then treated with each compound at a final concentration of 10 µM. After 24 hours of compound treatment, the degree of green luminescence was confirmed using GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA).
- For fluorescence intensity, C2C12-PHF20/YY1-promoter-GFP cells were dispensed at 4 × 105 in a 6-well culture plate and cultured for 24 hours. Then, cells in a 70% confluent state were treated with 250 ng/ml of doxycycline and then after 24 hours, treated with sulfamethazine, ciclopirox, or IOX1 for 24 hours at a final concentration of 10 µM, respectively. Thereafter, the degree of green luminescence was confirmed using GloMax microplate readers explorer (Promega, Madison, Wisconsin, USA).
- As a result, 13 kinds of compounds having an effect of reducing green luminescence were derived as candidate substances, in which the compounds included sulfasalazine, sulfamethazine, sulfathiazole, sulfapyridine, sulfaphenazole, sulfameter, sulfamethizole, sulfaguanidine, sulfacetamide sodium, sulfadoxin, sulfadimethoxine, sulfanilamide and sulfadiazine (
FIG. 4 ). - The effect was confirmed using one of the candidate substances, sulfamethazine. The sulfamethazine was purchased from Sigma (catalog No.S8876) and used, and then dissolved in DMSO at 10 mM and subsequently used in cell experiments.
- In order to quantitatively measure the inhibitory ability of myoblast formation inhibition according to a concentration of sulfamethazine, the HTS system constructed in Example 1 was treated with 1 nM, 10 nM, 100 nM, 1 µM or 10 µM of sulfamethazine, respectively, to measure green fluorescence.
- As a result, as illustrated in
FIG. 6 , it was confirmed that the higher the treated concentration of sulfamethazine, the weaker the intensity of fluorescence, which meant that the sulfamethazine acted on PHF20 to inhibit the expression of YY1 that inhibited the formation of myoblasts, and the inhibitory ability was improved in a concentration-dependent manner. - In addition, it was confirmed that the IC50 value of sulfamethazine was 0.591 µM based on the green fluorescence intensity obtained in the experiment (
FIG. 6 ). - In addition, the C2C12 muscle cell line was treated with each compound for each concentration for 24 hours, and the expression of proteins PHF20, YY1, and MyoD1 was confirmed by western blotting. In a group treated with each compound, it was confirmed that the protein expression of PHF20 and YY1 decreased, and the expression of MyoD increased (
FIG. 7 ). - A sterile coverslip was placed in a 12-well plate, and 1 × 105 C2C12/Tet-On PHF20 cells per well were plated and grown at 37°C. The cells were treated with doxycycline for 24 hours. After 24 hours, the cells were differentiated with DM (
HS 2%) for 5 days, and at this time, each sulfonamide-based compound was treated for each concentration onday 4 of differentiation. Thereafter, onday 5 of differentiation, the cells were washed twice with PBS at 37°C, reacted in 4% paraformaldehyde for 1 hour, and fixed on the coverslip. Then, the cells were washed twice with PBS. Before blocking, the cells were reacted with Triton x-100 in PBS for 30 minutes. The coverslip was blocked in 1% BSA for 1 hour at room temperature while shaking. An anti-Myosin (MF20) antibody was added to 1% BSA (1:200) and reacted overnight at 4 °C. Then, the coverslip was washed three times each with PBS, and an Alexa Fluor 568 secondary antibody (1:1000) in 1% BSA was added, and then reacted at room temperature in the dark for 1 hour while mixing. Thereafter, the coverslip was washed three times with PBS and mounted on a slide using a mounting medium containing DAPI VECTASHIELD (St. Louis, USA), and images of the stained cells were photographed using Zeiss, which were illustrated inFIGS. 8A to 8C . - Specifically,
FIG. 8A shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells after differentiation into myotubes from a group of treating a C2C12-PHF20 inducible cell line with doxycyclin to overexpress PHF20 and a control group treated with no doxycyclin, treating drugs (sulfamethazine, sulfatiazole, sulfadiazine, sulfadoxin, sulfadimethoxine, sulfacetamide sodium, sulfaphenazole, and sulfameter) onday 4 of differentiation, and then staining with heavy chain of myosin II (MF20) involved in muscle function and development onday 5 of differentiation. -
FIG. 8A shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfamethazine, sulfatiazole, sulfadiazine, sulfadoxin, sulfadimethoxine, sulfacetamide sodium, sulfaphenazole, and sulfameter,FIG. 8B shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfamethazine, sulfapyridine, sulfanilamide, and sulfamethizole, andFIG. 8C shows a cellular immunofluorescence staining image for confirming the differentiation ability of muscle cells when treated with sulfasalazine for each concentration. - 32 48-week-old C57BL/6J male mice were obtained from KBSI and then bred under constant room temperature and 12 h night/day cycle for 2 weeks before the experiment, and the mice were fed with a standard rodent diet and freely taken with water.
- Treadmill adaptation was performed randomly for 3 days under conditions of
grade 0, speed 15 (cm/sec), 10 minutes, electrical stimulation 02 AM (defined as exhaustion when electrical stimulation occurred consecutive 4 times or more), and according to the treadmill result, mice with similar ability were distributed into each group and divided into 4 groups of 8 mice per group. - In order to confirm the treadmill exercise capacity according to a dose of sulfasalazine, the mice divided in 3-1 above were orally administered at 10:30 every day for 28 days with
Group 1 of PBS (Hyclone Dulbecco's Phaosphate Buffer saline Cat. SH30028.02),Group 2 of sulfasalazine (Sigma, Cat. Nr S0883. CAS Number 599-79-1) 5 mpk,Group 3 ofsulfasalazine 50 mpk, andGroup 4 ofsulfasalazine 500 mpk. In order to reduce an error on treadmill exercise ability, the mice were starved for 3 hours before measuring the treadmill, and then the treadmill was performed 4 times at 7-day intervals (0 days, 7 days, 14 days, and 28 days) according to Table 1 below.[Table 1] Grade Time (min) Speed (cm/sec) 0 0 ∼ 3 15 0 3 ∼ 6 20 0 6 ∼ 9 25 0 9 ∼ 12 30 0 12 ∼ 15 35 0 15 ∼ 18 40 - Electrical stimulation 02 AM - Day 0 before administration,days - Defined as exhaustion when electric stimulation occurred consecutive 4 or more times - For a total of 28 days, the body weight of the mouse was measured, and any change in body weight was confirmed. As shown in
FIG. 9 , no difference in body weight was observed for each group during the entire experimental period. - As a result of confirming the treadmill exercise capacity, as shown in
FIG. 10 , it was confirmed that the higher the sulfasalazine dose, the higher the treadmill exercise capacity of the mouse ((∗) p ≤ 0.05 vs G1, (∗∗) p ≤ 0.01 vs G1 , (∗∗∗) p ≤ 0.001 vs G1). - In order to confirm the balance ability and the grip strength recovery effect of the Rota Rod according to a dose of sulfasalazine, 50-week-old C57BL/6J male mice were divided into
Group 1 of PBS,Group 2 ofsulfasalazine 5 mg/kg,Group 3 ofsulfasalazine 50 mg/kg, andGroup 4 ofsulfasalazine 500 mg/kg, and orally administered every day at 10 am for 28 days, and then the body weight was measured. - The Rota Rod was measured on
days Group 2 under the conditions shown in Table 2 below, and the results were shown inFIG. 11 ((∗) p ≤ 0.05 vs G1). In the same animal group as the Rota Rod, the grip strength was measured on the 2 paws and 4 paws of the mouse ondays FIGS. 12A and 12B .[Table 2] Constant test Acceleration 4 rpm in 30 second (mouse standing) 4-40 rpm in 300 second (until mouse fall down) - Wash with 50%EtOH - Interval 15 min - As shown in FIGS. 11A and 11B, it was confirmed that as the dose of sulfasalazine increased and the administration day increased, the recovery of balance ability of the mouse on the Rota Rod was improved. In addition, as shown in
FIGS. 12A and 12B , in both 2 paws and 4 paws measurement, it was confirmed that the grip strength was improved with the increase in sulfasalazine dose compared to a control group,Group 1, and the grip strength was improved as the administration day was increased. - In order to construct a high-fat diet CTX-induced sarcopenia mouse model, for 23-week-old C57BL/6J male mice subjected to 60% HFD for 19 weeks, cardiotoxin (LATOXAN, Portes-lès-Valence, France) was dissolved at 1 mg/ml in PBS to make a high-concentration stock solution. For application to mice, the stock solution was diluted again in PBS (Hyclone Dulbecco's Phaosphate Buffer saline Cat. SH30028.02) to a final concentration of 0.03 mg/ml and used, and intramuscularly injected into both quadriceps muscles of the mouse using a 1 ml syringe (24G) to construct the high-fat diet CTX-induced sarcopenia mouse model.
- Five groups of
Group 1 of orally administering PBS to a high-fat diet mouse without injecting CTX,Group 2 of orally administering PBS to a high-fat diet mouse injected with CTX,Group 3 of orally administeringsulfasalazine 5 mg/kg to the high-fat diet mouse injected with CTX,Group 4 of orally administeringsulfasalazine 50 mg/kg to the high-fat diet mouse injected with CTX, andGroup 5 of orally administeringsulfasalazine 500 mg/kg to the high-fat diet mouse injected with CTX were divided and orally administered at 10 am every day for 14 days, and the body weights were measured and the results were shown inFIG. 13 . - As shown in
FIG. 13 , no difference in body weight was observed for each group during the entire experimental period. - The measurement before injecting CTX into the sarcopenia mouse constructed in 4-1 above was set as
day 0, and ondays FIG. 14 . The measurement before injecting CTX into the same animal group as the Rota Rod was set asday 0, and ondays FIGS. 15A and 15B ((∗) p ≤ 0.05 vs G2).[Table 3] Constant test Acceleration 2 rpm in 30 second (mouse standing) 2-20 rpm in 300 second (until mouse fall down) - Wash with 50%EtOH - Interval 15 min - As shown in
FIG. 14 , it was confirmed that as the dose of sulfasalazine increased and the administration day increased as compared to a group injected with CTX, the recovery of balance ability of the mouse on the Rota Rod was improved. - In addition, as shown in
FIGS. 15A and 15B , in both 2 paws and 4 paws measurement, it was confirmed that the grip strength was improved with the increase in sulfasalazine dose compared to the group injected with CTX, and the grip strength was improved as the administration day was increased. - The left leg of an 8-week-old C57BL/6J male mouse was anesthetized with isoflurane, the entire leg was wound from the ankle with a 5 cm sports tape, and then wound and fixed with a Velcro with a width of 10 mm, and the Velcro condition was checked every day for 14 days (
FIG. 16 ). Meanwhile, in the present disclosure, Velcro injury means that muscle damage is caused by such Velcro by winding the muscle with the Velcro so as not to move. - In order to confirm the amount of treadmill exercise according to the dose of sulfasalazine, Group 1 (n=5) of orally administering PBS to 8-week-old C57BL/6J male mice without injury, Group 2 (n=5) of orally administering PBS to mice with Velcro injury, Group 3 (n=5) of orally administering
sulfasalazine 5 mg/kg to mice with Velcro injury, Group 4 (n=5) of orally administeringsulfasalazine 50 mg/kg to mice with Velcro injury, and Group 5 (n=5) of orally administeringsulfasalazine 500 mg/kg to mice with Velcro injury were divided and orally administered at 10 am every day for 14 days, and the body weights were measured and the results were illustrated inFIG. 18 . After 14 days, the Velcro was unwound, the treadmill was measured ondays [Table 4] Grade (%) Time (min) Speed (cm/sec) 0 0 ∼ 2 15 0 2 ∼ 3 25 0 3 ∼ 4 26 0 4 ∼ 5 27 0 5 ∼ 6 28 0 6 ∼ 7 29 Electrical stimulation 02 AM - Inclination 10°- Continue to increase until exhaustion at 1 cm/sec per minute - Day 0 before administration,days - Defined as exhaustion when electrical stimulation occurred for 5 seconds - For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed.
- As shown in
FIG. 17 , no difference in body weight was observed for each group during the entire experimental period. - In order to confirm the treadmill exercise capacity of mice with Velcro injury and mice without Velcro injury, as illustrated in
FIG. 18A , as a result of comparing distances run on the treadmill on the first day after unwinding the Velcro, it was confirmed that the mice wound with Velcro were injured with Velcro equally in each group (Group 2 to Group 5) compared to the mice unwound with Velcro (Group 1). As illustrated inFIG. 18B , the same result as the treadmill running distance was confirmed as compared with the treadmill running time. - In addition, as illustrated in
FIG. 19A , as a result of comparing the treadmill running distances on the day of unwinding the Velcro, anddays day 6 of administration compared to a comparative group without winding Velcro, and the treadmill running distances were increased as the administration period was increased. - In addition, as illustrated in
FIG. 19B , as a result of comparing the treadmill running distances on the day of unwinding the Velcro, anddays day 6 of administration compared to a comparative group without winding Velcro, and the treadmill running time was increased as the administration period was increased ((∗) p ≤ 0.05, (∗∗) p ≤ 0.01, (∗∗∗) p ≤ 0.001). - As experimental animals, 7-week-old male and female C57BL/6J mice with the body weight of 20 ± 3 g were obtained from DooYeol Biotech (Seoul, South Korea). The mice were bred under constant room temperature and 12 h night/day cycle for 1 week before the experiment, and the mice were fed with a standard rodent diet and were freely taken with water.
- In order to construct a CTX-induced sarcopenia mouse model, cardiotoxin (LATOXAN, Portes-lès-Valence, France) was dissolved at 1 mg/ml in PBS to make a high-concentration stock solution. To apply to the mice, the stock solution was diluted again in PBS at a final concentration of 0.03 mg/ml and used, and intramuscularly injected into both thigh muscles of the mouse using a 1 ml syringe (24G) to construct a sarcopenia mouse model.
- Thereafter, in an experiment for confirming the effect of the sulfonamide-based compounds of the present disclosure, the efficacy was confirmed using the CTX-induced sarcopenia mouse model.
- 4 µM of sulfamethazine was orally administered to the mouse model of 6-1 (after 24 hours of CTX administration), the muscle tissue was collected from mice on
days - Specifically, for mouse muscle tissue immunostaining, the muscle tissues of a comparative group and a sulfamethazine-treated group were fixed in 4% paraformaldehyde, washed, and fixed in paraffin. Tissues were cut to a thickness of about 4 mm, fixed to a slide, and then stained using an antibody for each muscle type.
- As illustrated in
FIG. 20 , as a result of confirming the degree of muscle differentiation of the muscle tissue of the mouse through H&E staining, in a control group (a non-sulfamethazine administered group), it was confirmed that the muscle were damaged by CTX, and in a group orally administered with sulfamethazine, it was confirmed that the muscle was regenerated compared to the control group. In particular, after treatment with sulfamethazine, it may be seen that muscle regeneration occurs fromday 3, and muscle regeneration occurs quickly ondays - In the same manner as in 6-2 above, 4 µM of sulfamethazine was administered to the mouse, and on
days - As illustrated in
FIG. 21 , in the sulfamethazine-treated group, compared to the control group, an increase in expressions of green myosin IIa and red myosin IIb was confirmed, and onday 6, the expression of blue-stained myosin I was increased, and the image onday 10 was observed to be recovered similarly to that of a normal control group onday 0. This means that the recovery of muscle damage in the sulfamethazine-administered group is faster than that of a CTX-induced muscle loss mouse (the non-sulfamethazine administered group). - In order to confirm the balance ability according to a dose of sulfamethazine, 7-week-old male mice were divided into groups of 10 mice to set experimental groups as shown in Table 5, and the experiment was conducted. Specifically, the Rota Rod was measured before CTX injection, and after CTX was injected (day 0), the drug was orally administered once/day for 10 days, and on
days FIG. 23 .[Table 5] (-)PBS group (+) PBS group 50 mpk group 100 mpk group 500 mpk group CTX-administration X O O O O Sulfamethazine PBS PBS 50 mg/ kg 100 mg/ kg 500 mg/kg - CTX-administration: 0.03 mg/ml, 50 µl injection to each of both hind legs - Administration period of sulfamethazine: For 10 days, once/day, oral administration - Rota Rod measurement: Before CTX injection, and 4 days, 9 days, and 14 days after drug administration - 10 mice per each group - Sulfamethazine: Thermo Fisher, Cat. A19276. CAS Number 57-68-1 - Administration: DMSO 10% and Cremorphor 12.5% were mixed and then diluted with PBS and administered - For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed. As illustrated in
FIG. 22 , no change in body weight was observed for each group during the entire experimental period, except for a decrease in body weight in all groups onday 10 without diet supply. - As a result of confirming the balance ability of the Rota Rod, as illustrated in FIGS. 24A and 24B, compared to a PBS-administered group ((+)PBS group) after CTX injection, it was confirmed that the higher the dose of sulfamethazine, the higher the balance ability of the Rota Rod of the mouse. In the case of a group without inducing muscle loss ((-)PBS group), the improvement in the balance ability of the Rota Rod according to repeated exercise was observed, but in the case of a sulfametazine-administered group, the balance ability was improved in a dose-dependent manner.
- In order to confirm the grip strength recovery according to a dose of sulfamethazine, 7-week-old male mice were divided into groups of 10 mice to set experimental groups as shown in Table 6, and the experiment was conducted. Specifically, the grip strength was measured before CTX injection, and after CTX was injected (day 0), the drug was orally administered once/day for 10 days, and on
days [Table 6] (-)PB S group (+) PB S group 50 mpk group 100 mpk group 500 mpk group CTX-injection X O O O O Sulfamethazine PBS PBS 50 mg/ kg 100 mg/ kg 500 mg/kg - CTX-administration: 0.03 mg/ml, 50 µl injection to each of both hind legs - Administration period of sulfamethazine: For 10 days, once/day, oral administration - Rota Rod measurement: Before CTX injection, and 4 days, 9 days, and 14 days after drug administration - 10 mice per each group - Sulfamethazine: Thermo Fisher, Cat. A19276. CAS Number 57-68-1 - Administration: DMSO 10% and Cremorphor 12.5% were mixed and then diluted with PBS and administered - For a total of 14 days, the body weights of the mice were measured, and any change in body weight was confirmed. As illustrated in
FIG. 22 , no change in body weight was observed for each group during the entire experimental period, except for a decrease in body weight in all groups onday 10 without diet supply. - As a result of confirming the grip strength recovery effect, as shown in
FIGS. 26A to 26D, for the exercise capacity of the hind legs (2 paws) injected with CTX, compared to the PBS-administered group ((+)PBS group) after CTX injection, it was confirmed that the grip strength of mice increased as the dose of sulfamethazine increased. As a result of measuring the grip strength of all four paws, it was also showed that the grip strength was increased in a sulfamethazine concentration-dependent manner. Similarly to the group ((-)PBS group) without inducing muscle loss on the Rota Rod, which had showed a repeated level of improvement in balance ability, even in the grip strength, it was confirmed that the grip strength recovery was improved in a dose-dependent manner in the sulfamethazine-administered group. - 7-week-old male and female C57BL/6J mice with the body weight of 20 ± 3 g were obtained from DooYeol Biotech (Seoul, South Korea). The mice were bred under constant room temperature and 12 h night/day cycle for 1 week before the experiment, and the mice were fed with a standard rodent diet and were freely taken with water. The experiment was conducted by dividing the mice into a control group and test groups (100 mg/kg, 500 mg/kg, and 1000 mg/kg), and sulfamethazine was diluted with 12.5% DMSO and 12.5% cremophor and orally administered at 10 ml/kg.
- Control group: PBS (without administration of sulfamethazine)
- Group 1: 100 mg/kg (experimental group, administration of sulfamethazine)
- Group 2: 500 mg/kg (experimental group, administration of sulfamethazine)
- Group 3: 1,000 mg/kg (experimental group, administration of sulfamethazine)
- The experiment was conducted for 2 weeks, and the general condition and the presence or absence of death were observed at least once a day. The body weights were measured before administration, and on 1 day, 3 days, 7 days, and 14 days after administration, and an autopsy was performed on
day 14. - In the present disclosure, a single oral dose toxicity test refers to a test that qualitatively and quantitatively examines toxicity in a short period of time when a test substance was administered to test animals in a single dose (including divided doses within 24 hours).
- Specifically,
FIG. 27A shows all mice for confirming toxicity according to single oral administration of female and male mice,FIG. 27B shows photographs of male mice before organ harvesting, andFIG. 27C shows photographs of female mice before organ harvesting. - In addition, all organs of the head, the thoracic cavity, and the abdominal cavity were observed, which were shown in
FIGS. 28A to 39 . - Specifically,
FIG. 28A is a photograph confirming toxicity by dissecting the livers of male mice,FIG. 28B is a photograph confirming toxicity by dissecting the livers of female mice,FIG. 29A is a photograph confirming toxicity by dissecting the lungs of male mice, andFIG. 29B is a photograph confirming toxicity by dissecting the lungs of female mice.FIG. 30 is a photograph of confirming toxicity by dissecting the brains of male and female mice,FIG. 31 is a photograph of confirming toxicity by dissecting the hearts of male and female mice,FIG. 32 is a photograph of confirming toxicity by dissecting the stomachs of male and female mice,FIG. 33 is a photograph of confirming toxicity by dissecting the pancreases of male and female mice, andFIG. 34 is a photograph of confirming toxicity by dissecting the spleens of male and female mice.FIG. 35A is a photograph of confirming toxicity by dissecting the kidneys of male mice,FIG. 35B is a photograph of confirming toxicity by dissecting the kidneys of female mice,FIG. 36A is a photograph of confirming toxicity by dissecting the S. intestines of male mice, andFIG. 36B is a photograph of confirming toxicity by dissecting the S. intestines of female mice.FIG. 37A is a photograph of confirming toxicity by dissecting the L. intestines of male mice,FIG. 37B is a photograph of confirming toxicity by dissecting the L. intestines of female mice,FIG. 38 is a photograph of confirming toxicity by dissecting the testis of male mice, andFIG. 39 is a photograph of confirming toxicity by dissecting the wombs of female mice. - In addition, after oral administration of the PBS or sulfamethazine, the results of measuring the body weights of male mice and female mice in each group on
days FIG. 40 , the results of measuring the weight of each tissue of each male mouse onday 14 were shown inFIG. 41A , and the results of measuring the weight of each tissue of each female mouse onday 14 were shown inFIG. 41B . There was no singularity in tissue weight for each group, and it was confirmed that the decrease in pancreas weights of females in the 500 mpk group was caused by the loss of some tissues during dissection. - As in Example 7-1, mice with single oral administration were dissected on
day 14 after administration, and histochemical toxicity was evaluated. The pathology of the liver was observed through H&E staining on the dissected liver tissue. - As shown in
FIGS. 42A and42B , no specific pathological findings were observed between each group, similarly to the fact that the toxicity was not confirmed in the harvested livers of male and female mice. - In mice orally administered once as in Experimental Example 7-1, blood was collected from the heart of the mouse on
day 14 after administration, and serum was collected to confirm the expression of a liver toxicity marker, a heart toxicity marker, and a kidney toxicity marker, respectively. Specifically, blood samples were collected from the heart after surgery onday 14 after administration of sulfamethazine. The collected blood was left at room temperature for 1 hour to be coagulated, and the serum was isolated by operating a centrifuge (13,000 rpm) for 20 minutes. Blood analysis was performed by dividing liver toxicity, heart toxicity, and kidney toxicity, and the analysis was conducted by requesting DooYeol Biotech (Seoul, Korea). - As a result, as shown in
FIGS. 43A and43B , no specific phenomena related to liver toxicity, heart toxicity, and kidney toxicity in male and female mice were observed in the sulfamethazine-administered group. - From the foregoing, it will be appreciated that various embodiments of the present disclosure have been described herein for purposes of illustration, and that various modifications may be made without departing from the scope and spirit of the present disclosure. Accordingly, the various embodiments disclosed herein are not intended to be limiting, with the true scope and spirit being indicated by the following claims.
Claims (11)
- A pharmaceutical composition for use in prevention or treatment of a muscular disorders, the compositon comprising a sulfonamide-based compound represented by Chemical Formula 1 below or a pharmaceutically acceptable salt thereof:
- The pharmaceutical composition of claim 1, wherein the aryl or heteroaryl includes thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine, and the aryl or heteroaryl is unsubstituted, or substituted with at least one substituent selected from C1-3 alkyl, C1-3 alkoxy, and phenyl.
- The pharmaceutical composition of claim 1 or 2, wherein the at least one substituent selected from C1-3 alkyl and C1-3 alkoxy is methyl or methoxy.
- The pharmaceutical composition of claim 1, wherein the muscular disorder is any one selected from the group consisting of sarcopenia, atony, muscular atrophy, muscular dystrophy, muscle degeneration, cachexia and myasthenia.
- A food composition for use in prevention or improvement of a muscular disorders, the composition comprising a sulfonamide-based compound represented by Chemical Formula 1 below or a food acceptable salt thereof:
- The food composition of claim 6, wherein the aryl or heteroaryl includes thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine, and the aryl or heteroaryl is unsubstituted, or substituted with at least one substituent selected from C1-3 alkyl, C1-3 alkoxy, and phenyl.
- The food composition of claim 6 or 7, wherein the at least one substituent selected from C1-3 alkyl and C1-3 alkoxy is methyl or methoxy.
- A feed composition for use in prevention or improvement of a muscular disorders, the composition comprising a sulfonamide-based compound represented by Chemical Formula 1 below or a salt thereof:
- The feed composition of claim 9, wherein the aryl or heteroaryl includes thiazole, diazole, thiadiazole, phenyl, pyridine or pyrimidine, and the aryl or heteroaryl is unsubstituted, or substituted with at least one substituent selected from C1-3 alkyl, C1-3 alkoxy, and phenyl.
- The feed composition of claim 9 or 10, wherein the at least one substituent selected from C1-3 alkyl and C1-3 alkoxy is methyl or methoxy.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR20220043350 | 2022-04-07 | ||
KR20220058865 | 2022-05-13 | ||
KR1020220186355A KR20230144932A (en) | 2022-04-07 | 2022-12-27 | Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds |
Publications (2)
Publication Number | Publication Date |
---|---|
EP4257139A2 true EP4257139A2 (en) | 2023-10-11 |
EP4257139A3 EP4257139A3 (en) | 2023-12-13 |
Family
ID=85980468
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP23166953.2A Pending EP4257139A3 (en) | 2022-04-07 | 2023-04-06 | Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds |
Country Status (3)
Country | Link |
---|---|
US (1) | US20230321096A1 (en) |
EP (1) | EP4257139A3 (en) |
JP (1) | JP2023155212A (en) |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DE69232909D1 (en) * | 1991-02-22 | 2003-03-06 | Howard K Shapiro | USE OF PHARMACEUTICAL ACTIVE SUBSTANCES FOR THE TREATMENT OF DISEASE SYMPTOMS, NEUROLOGICAL DISEASES AND ETIOLOGICALLY RELATED SYMPTOMS COMPLEXES |
US20100292306A1 (en) * | 2005-05-24 | 2010-11-18 | Carlson C George | Compositions And Methods For The Treatment Of Muscular Dystrophy |
CA2613337A1 (en) * | 2005-05-24 | 2006-11-30 | A. T. Still University Of Health Sciences | Nk-b inhibitors for the treatment of muscular dystrophy |
UA113165C2 (en) * | 2011-03-01 | 2016-12-26 | APPLICATION OF A COMBINATION OF Baclofen AND ACOMPROSAT FOR THE TREATMENT OF NEUROLOGICAL DISEASES AND A COMPOSITION CONTAINING Baclofen AND ACAMPROSAT | |
WO2016061509A1 (en) * | 2014-10-17 | 2016-04-21 | The Broad Institute, Inc. | Compositions and methods of treatng muscular dystrophy |
KR20160069802A (en) * | 2014-12-09 | 2016-06-17 | 삼성전자주식회사 | Composition for preventing or treating muscular atrophy or muscular regeneration in a subject comprising a sulfonamide compound and use thereof |
-
2023
- 2023-04-06 JP JP2023062388A patent/JP2023155212A/en active Pending
- 2023-04-06 EP EP23166953.2A patent/EP4257139A3/en active Pending
- 2023-04-06 US US18/296,796 patent/US20230321096A1/en active Pending
Non-Patent Citations (1)
Title |
---|
CAS, no. 599-79-1 |
Also Published As
Publication number | Publication date |
---|---|
EP4257139A3 (en) | 2023-12-13 |
US20230321096A1 (en) | 2023-10-12 |
JP2023155212A (en) | 2023-10-20 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10646498B2 (en) | Composition for preventing, alleviating or treating muscle diseases or improving muscular function | |
CN101484158B (en) | Senescence inhibitor | |
US20220241229A1 (en) | Methods and Compositions for Altering Senescence Associated Secretory Phenotype | |
US20090306208A1 (en) | Prophylactic or therapeutic composition for hemoglobinuria or myoglobinuria | |
KR20210014231A (en) | Composition for prevention or treatment of muscular disorders or improvement of muscular functions comprising seaweeds extract | |
JP7149548B2 (en) | Composition for prevention, improvement or treatment of muscle diseases, or for improving muscle function, containing an extract of damselfly | |
KR101842948B1 (en) | Composition comprising Decanal or as active ingredients for Preventing or treating muscle disease | |
EP4257139A2 (en) | Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds | |
WO2011145909A2 (en) | Novel use of panduratin derivatives or an extract of boesenbergia pandurata for promoting muscle mass growth, fighting fatigue, and enhancing exercise performance capability | |
WO2020218148A1 (en) | Lipid peroxide production inhibitor | |
KR102159019B1 (en) | Composition for preventing or treating obesity comprising Honeybee Drone Pupas | |
KR20230144932A (en) | Composition for improving, preventing or treating muscular disorders comprising sulfonamide-based compounds | |
KR20200083146A (en) | Composition for prevention and treatment of muscular disorder or improvement of muscular functions comprising Illicium verum extract or shikimic acid | |
KR102352636B1 (en) | Composition for prevention and treatment of muscular disorder or improvement of muscular functions comprising Illicium verum extract or shikimic acid | |
CN116889569A (en) | Composition for improving, treating or preventing muscle diseases comprising sulfonamide compound | |
KR101121954B1 (en) | Composition for preventing or treating diabetes comprising 1,2,3-Benzentricarboxylic acid | |
US20240293493A1 (en) | Composition for alleviating, treating, or preventing muscular diseases, or improving muscular functions, containing korean mint extract or tilianin as active ingredient | |
KR102517662B1 (en) | Composition for improvement, treatment or prevention of muscular disorders, or improvement of muscular functions comprising chaga | |
KR102380295B1 (en) | A composition for preventing, improving or treating sarcopenia comprising extracts of oat | |
KR102421411B1 (en) | Composition for preventing or treating Muscular Dystrophy comprising Oxiracetam | |
US20220088106A1 (en) | Composition comprising cudrania tricuspidate as effective component for alleviating, treating, or preventing muscular diseases, or improving muscule functions | |
WO2022030719A1 (en) | Composition for improving, treating, or preventing muscle disease, or improving muscle function, containing rose hip as acitve ingredient | |
KR20240029547A (en) | Composition for improvement, prevention or treatment of muscular disorders, or improvement of muscular functions comprising quinic acid | |
KR20240133872A (en) | Composition for improvement, treatment or prevention of muscular disorders, or improvement of muscular functions comprising rye pollen | |
EP4331579A1 (en) | Composition comprising oxiracetam for preventing or treating muscular disease |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC ME MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
PUAL | Search report despatched |
Free format text: ORIGINAL CODE: 0009013 |
|
AK | Designated contracting states |
Kind code of ref document: A3 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC ME MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
RIC1 | Information provided on ipc code assigned before grant |
Ipc: A61P 21/00 20060101ALI20231108BHEP Ipc: A61K 31/635 20060101ALI20231108BHEP Ipc: A61K 31/63 20060101AFI20231108BHEP |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE |
|
17P | Request for examination filed |
Effective date: 20240613 |
|
RBV | Designated contracting states (corrected) |
Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC ME MK MT NL NO PL PT RO RS SE SI SK SM TR |